Next Article in Journal
Combined Application of Organic Fertilizer with Microbial Inoculum Improved Aggregate Formation and Salt Leaching in a Secondary Salinized Soil
Next Article in Special Issue
An In Vitro Evaluation and Network Pharmacology Analysis of Prospective Anti-Prostate Cancer Activity from Perilla frutescens
Previous Article in Journal
Chemical Constituents with Anti-Lipid Droplet Accumulation and Anti-Inflammatory Activity from Elaeagnus glabra
Previous Article in Special Issue
From Plants to Wound Dressing and Transdermal Delivery of Bioactive Compounds
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects

Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
*
Author to whom correspondence should be addressed.
Plants 2023, 12(16), 2944; https://doi.org/10.3390/plants12162944
Submission received: 17 July 2023 / Revised: 11 August 2023 / Accepted: 13 August 2023 / Published: 14 August 2023

Abstract

:
The rising predominance of type 2 diabetes, combined with the poor medical effects seen with commercially available anti-diabetic medications, has motivated the development of innovative treatment approaches for regulating postprandial glucose levels. Natural carbohydrate digestion enzyme inhibitors might be a viable option for blocking dietary carbohydrate absorption with fewer side effects than manufactured medicines. Alpha-amylase is a metalloenzyme that facilitates digestion by breaking down polysaccharides into smaller molecules such as maltose and maltotriose. It also contributes to elevated blood glucose levels and postprandial hyperglycemia. As a result, scientists are being urged to target α-amylase and create inhibitors that can slow down the release of glucose from carbohydrate chains and prolong its absorption, thereby resulting in lower postprandial plasma glucose levels. Natural α-amylase inhibitors derived from plants have gained popularity as safe and cost-effective alternatives. The bioactive components responsible for the inhibitory actions of various plant extracts have been identified through phytochemical research, paving the way for further development and application. The majority of the findings, however, are based on in vitro investigations. Only a few animal experiments and very few human investigations have confirmed these findings. Despite some promising results, additional investigation is needed to develop feasible anti-diabetic drugs based on plant-derived pancreatic α-amylase inhibitors. This review summarizes the most recent findings from research on plant-derived pancreatic α-amylase inhibitors, including plant extracts and plant-derived bioactive compounds. Furthermore, it offers insights into the structural aspects of the crucial therapeutic target, α-amylases, in addition to their interactions with inhibitors.

1. Introduction

Diabetes mellitus is a group of metabolic disorders marked by persistent increases in blood glucose levels. This condition is caused by a defect in insulin production, insulin function, or both factors. Insulin insufficiency and its resistance to the intended target tissues cause abnormalities in the metabolic processing of carbohydrates, lipids, and proteins [1,2,3]. Diabetes is linked to several short- and long-term health ramifications. Acute health problems include diabetic ketoacidosis, malignant hyperthermia-like syndrome with rhabdomyolysis, and hyperosmolar hyperglycemia, all of which carry a significant risk of morbidity and fatality in the near term [4,5]. The long-term ramifications might entail hypertension, lipid disorders, retinopathy, renal malfunction, nonalcoholic fatty liver disease, neurological diseases, cardiovascular and atherosclerotic problems, and others [4,5]. The most prevalent signs of hyperglycemia include polyuria, excessive thirst, unexplained weight loss, hyperphagia, and a lack of visual acuity [6]. Depending on the mode of its manifestation, there are several different types of diabetes, which include: (1) type 1 diabetes (T1D), which is caused by the destruction of pancreatic β-cells by the body’s immune system; (2) type 2 diabetes (T2D), which is characterized primarily by reduced insulin receptor sensitivity; (3) gestational diabetes (GD), which is recognized in the second or third trimester of pregnancy but was not concisely identified as diabetes prior to gestation; and (4) particular kinds of diabetes caused by different factors [7]. Diabetes mellitus is a major global disease, affecting over half a billion individuals in 2021, and the number of adults affected is expected to reach nearly 800 million by 2045 [8,9]. T2D, which is the most prevalent type (~90%), is a metabolic disorder that involves carbohydrate, lipid, and protein metabolic abnormalities as well as deficiencies in insulin secretion, typically accompanied by insulin resistance [10]. These chronic metabolic disorders may result in neuropathy, retinopathy, angiopathy, and nephropathy [11]. In this perspective, carbohydrate-digesting enzyme inhibition is regarded as a therapeutic strategy for the management and treatment of T2D. The most abundant carbohydrate in meals is starch, which is composed of two distinguished polysaccharides: one is linear with α-(1→4) glycosidic bonds (amylose), and the other is branched with α-(1→6) glycosidic bonds (amylopectin) [12]. Pancreatic α-amylase (EC 3.2.1.1) is the most essential digestive enzyme. It is a calcium-based metalloenzyme that acts as a catalyst and facilitates the hydrolysis of the α-1,4 glycosidic bonds of polysaccharide molecules such as amylose, amylopectin, glycogen, and other maltodextrins and is accountable for the majority of starch digestion in humans. Another digestive enzyme, α-glucosidase or maltase (EC 3.2.1.20), catalyzes the final phase of carbohydrate digestion, acting on 1,4-alpha bonds to generate glucose (Figure 1) [13].
Scientific investigations have shown the association between the activities of human pancreatic α-amylase (HPA) and postprandial glucose levels, highlighting the importance of lowering postprandial hyperglycemia (PPHG) in the management of T2D [14]. The capacity of α-amylase enzyme inhibitors to prevent the digestion and absorption of dietary starch has led to their being categorized as starch blockers. Nevertheless, α-pancreatic amylase activity inhibition should be moderated to avoid bacterial fermentation of non-digested carbohydrates in the colon caused by excessive suppression of the activity of this enzyme, which gives rise to diarrhea and flatulence [15]. There are already certain diabetes medications that work primarily by limiting carbohydrate digestion and absorption. The first α-glucosidase inhibitor accessible for diabetic therapy was acarbose, a microbial inhibitor that inhibits α-amylase, maltase, and sucrase (EC 3.2.1.48) activities. Voglibose is a novel bacterial α-glucosidase inhibitor that suppresses maltase, isomaltase (EC 3.2.1.10), and sucrase activities, whereas miglitol is a 1-deoxynojirimycin derivative that functions by inhibiting the activities of glucoamylase (EC 3.2.1.3), sucrase, and isomaltase [16,17]. Although these medicines are effective in stabilizing postprandial blood glucose levels in a large number of individuals, they are frequently linked with substantial gastrointestinal side effects [16]. Furthermore, the undesirable clinical outcomes associated with marketed anti-hyperglycemic drugs are partly to blame for the common medication failure to comply that occurs in diabetes patients [18]. Because of the significant side effects of such medications, researchers have been looking for substitute therapies with minimal or no harmful effects. Although these anti-diabetic medicines are commercially available, several attempts have been made to produce non-cytotoxic anti-diabetic synthetic molecules [19,20,21]. From such a perspective, herbal chemicals intend to offer milder techniques of controlling metabolic problems and have been employed in traditional medical systems such as Indian Ayurveda (alternative medicine), Chinese herbal medicines, and Arabic Unani (traditional medicine used by the Muslims) since ancient times [22]. As a result, there is evidence suggesting the potentially helpful impact of a vast variety of medicinal herbs in T2D management [23]. Aside from their efficiency, herbal therapies appear to have few side effects and offer a cost-effective alternative to ingested commercial hypoglycemic medications. The World Health Organization (WHO) suggested in 1990 that extensive studies be conducted on the positive benefits of these plants [24]. In this regard, the current review highlights research on the α-amylase inhibitory action of plants and their phytochemical components, which may be effective in the treatment of diabetes. The review was conducted utilizing a scientific database composed of web search engines, including SciFinder, Scopus, Google Scholar, PubMed, and Science Direct, to examine published material between 2019 and 2023. Search criteria included ‘amylase inhibitor’, ‘anti-diabetic characteristics’, ‘plant extracts’, and ‘pancreatic amylase’. The latest articles related to pancreatic amylase (porcine and human) inhibition by plant extracts were also included. Medical plants having a folkloric background and α-amylase activity met the inclusion requirements.

2. Alpha-Amylase Structure and Mechanism of Action

In the human diet, starch serves as the main energy source. Dietary sugars and starch are broken down to glucose by α-glucosidase and α-amylase enzymes. α-amylase metalloenzymes can be found in the saliva and pancreatic juice and are members of the glycoside hydrolase family 13 (GH13) [25,26,27,28]. Metalloenzymes are a diverse collection of enzymes that utilize a metal cation as a co-factor in the enzyme’s active site. These enzymes stimulate a wide range of reactions, including hydrolytic activities [29]. α-amylase is a metalloenzyme that needs an important calcium ion for structural integrity and is activated by chloride ions [30,31,32]. Although this family’s overall amino acid sequence homology is low, it has short regions of highly conserved residues, and structural investigations have shown that its members do have comparable three-dimensional structures [33]. These isozymes are members of a multigene family located on chromosome 1 that is controlled such that the various isozymes are only expressed in the pancreas or salivary glands [34]. The genes AMY1 and AMY2 produce both salivary and pancreatic α-amylase isozymes, each of which has 496 amino acids in one polypeptide chain [35,36,37]. Before starch is absorbed, salivary and pancreatic α-amylases hydrolyze it in the mouth and small intestine [25,38,39]. Through the cleavage of 1-4-α-glycosidic bonds, these isozymes hydrolyze carbohydrate polymers into shorter oligomers, such as maltose, a-limit dextrins, and maltotriose. An early partial cleavage into smaller oligomers (10–30%) is provided by the salivary isozyme [37,40]. When partially digested saccharides enter the gut, pancreatic amylase, which is produced in the pancreas and secreted into the lumen, extensively hydrolyzes them into smaller oligosaccharides [38]. Subsequently, α-glucosidases located in the brush border hydrolyze the α-amylase products further into glucose in the lumen of the small intestine [25,39,41,42]. Afterward, glucose transporters take the glucose from the intestinal mucosa and transport it into the blood circulation [42].
The 3D structures of the α-amylase enzymes from human saliva, pancreas, and pig pancreas have been determined using X-ray crystallography [33,43,44]. These enzymes’ architectures are all quite similar to one another. Three structural domains (A, B, and C) make up mammalian amylases, the biggest of which, Domain A, creates a standard core of (β-α)8 barrel fold (Figure 2a), one end of which is positioned at the crucial active site residues (the catalytic triad of two aspartate (D197 and D300) and one glutamate (E233) residue) (Figure 3b). The active site is located in a substantial cleft that separates the carboxyl termini of the A and B domains. A bound chloride ion is present in Domain A as well, and it has been long recognized to activate amylase [32,45]. The bound chloride ion was found to form ligand interactions with R195, N298, and R337 in close proximity to the active site (Figure 2c) [33]. Domain B, the smallest domain, creates a calcium-binding site against Domain A’s β-barrels. The bound Ca2+ ion in Domain B, which also borders the area of the active site, is likely crucial in preserving the active site region protein configuration. N100, R158, D167, and H201, which make ligand interactions with calcium, may greatly contribute to its function (Figure 2b). Anti-parallel β-structure makes up Domain C, which is only tangentially related to Domains A and B. Notably, a stable pyrrolidone derivative is created by post-translational alteration of human pancreatic a-amylase’s N-terminal glutamate residue, which may offer defense against other digesting enzymes. It does not seem plausible that the molecule’s N-terminal Domain C will directly contribute to the catalytic process as it has a weaker connection to Domains A and B [33,46].
The catalytic processes in amylases are excellent representatives of a twofold displacement mechanism, in which the carboxyl groups of Asp and Glu residues serve as acid-base catalysts and a nucleophilic reactant in the creation of a covalent intermediate during the catalysis sequence. Through the use of a charge relay mechanism, a Cl ion may facilitate the protonation of a catalytically significant carboxyl group, leading to the activation [49,50,51]. With maltotriose and maltose as the main short oligomer yields (very little production of glucose), α-amylase is an endoenzyme that attacks linear sections of amylose and amylopectin several times [52,53,54]. According to kinetic studies, the pig pancreatic enzyme’s active site may take up to five glucose molecules, meaning that there are five sub-sites where glucose molecules can bind [52]. Several three-dimensional structures demonstrated the presence of multiple sites, but the pig pancreatic amylase structure complexed with acarbose showed a sixth site [44]. The target of anti-diabetic drugs such as α-amylase and α-glucosidase inhibitors is to reduce postprandial hyperglycemia, with the most widely used ones being acarbose, voglibose, and miglitol. However, when used in therapy, the inhibitors of α-glucosidase and α-amylase have been associated with gastrointestinal adverse effects such as diarrhea, bloating, and flatulence. The quest for the development of novel α-amylase and α-glucosidase inhibitors is therefore crucial for the management of PPHG in T2D. Such insights into α-amylase’s structure can be used to better understand the interaction of α-amylase with various inhibitors in order to develop anti-diabetic medicines with fewer adverse effects.

3. Plant Extracts as an α-Amylase Inhibitor Source

Several traditional medicines, in addition to the existing therapeutic options, have been promoted for diabetes therapy. Traditional plant medicines are used to manage a broad variety of diabetic symptoms all around the world. It is well acknowledged that plant remedies have fewer negative effects than modern pharmaceuticals [55,56,57,58,59,60], as well as being less expensive, driving both the public and healthcare institutions to investigate natural medicinal goods as alternatives to synthetic medications. As a consequence, studies on traditional medicinal herb-derived substances have grown in importance [61,62]. Several folkloric/medicinal plant extracts have been shown to possess potent α-amylase inhibition activity; however, further animal studies are needed to confirm their hypoglycemic physiological impact. Many studies investigated the potential role of medicinal plants in inhibiting α-amylase enzymes (Table 1). Among the latest plant extracts investigated in the literature that are featured in this study, Prosopis cineraria (L.), Terfezia claveryi, Chenopodium album L., and Salvia lavandulifolia Vahl have the highest potential to inhibit α-amylase enzymes (Table 1).
A recent study investigated the hypoglycemic effect of edible plant leaves from Palestine that are used as folkloric anti-diabetic remedies [67]. The lipophilic and hydrophobic fractions of these plants were tested against the porcine pancreatic α-amylase enzyme. The hydrophilic fractions of Centaurea iberica and Cichorium endivia showed the highest α-amylase activity with IC50 values of 12.33 μg/mL and 9.96 μg/mL, respectively. Furthermore, the highest α-amylase inhibition effect for lipophilic fractions was observed for Sisymbrium irio and Arum palaestinum, with IC50 values of 7.72 μg/mL and 25.3 μg/mL, respectively. Additionally, rice extract’s biological activity against α-amylase was investigated in Thailand, and the study showed that brown rice extract as well as the rice’s volatile compounds, identified as vanillin and vanillyl alcohol, have high inhibitory effects against α-amylase [68]. It is notable to mention that a synergy effect was noticed on α-amylase inhibition activity when a combination of black, red, and white rice extracts along with vanillin and vanillyl alcohol was used.
Rhus coriaria L. leaves and fruits are an important folk medicine that is used in Turkey for the treatment of diabetes [104]. Gök et al. studied the effect of R. coriaria ethanol extracts of leaf and fruit on α-amylase inhibitory activity in an attempt to isolate the active compounds against α-amylase [70]. The ethyl sub-extract of R. coriaria showed good α-amylase inhibition activity with an IC50 of 20.81 μg/mL against 26.99 μg/mL for acarbose. The study showed the successful isolation of several compounds; among them, penta-O-galloyl-β-glucopyranose, one of the main compounds in leaf and fruit extracts, had α-amylase inhibition activity with an IC50 of 6.32 μM compared to 10.69 μM for acarbose. Peanuts (Arachis hypogaea), another edible food, were investigated for their α-amylase inhibition property [72]. Various organic extracts of peanuts were investigated, and it was found that peanut seed ethanol extract has an α-amylase suppressing ability with an IC50 of 0.61 μg/mL, close to 0.31 μg/mL for acarbose, and the least cytotoxicity with an LC50 of 413.9 μg/mL. Melilotus officinalis (yellow sweet clover) is a medicinal plant typically employed in Asia and Europe and has been used as an anti-inflammatory traditional medicine [105,106]. As M. officinalis contains a high amount of coumarin derivatives, its extracts were clinically tested for the treatment of diabetic foot [107]. Paun et al. studied the polyphenolic-rich extracts of M. officinalis and their anti-diabetic activity [76]. M. officinalis polyphenolic-rich extracts displayed notable α-amylase inhibitory activity with an IC50 of 1.30 μg/mL, while acarbose showed an IC50 of 17.68 µg/mL, suggesting that it could be a good candidate for developing a natural anti-diabetic food supplement.
Solanum species are a rich source of traditional medicine remedies as antipyretic, anti-inflammatory, antioxidant, and anti-diabetic agents [108,109,110]. Ju’a-açu fruit (Solanum oocarpum), also known as Brazilian sunberry, has an alkaloid composition and has been reported to have anti-diabetic activity [111]. Saraswathi et al. studied the phytoconstituents of the ethanolic and aqueous extracts of Solanum virginianum dried fruits for their anti-diabetic activity [92]. The study showed that both the ethanolic and aqueous fruit extracts have α-amylase inhibition activity, with the aqueous extract having higher enzymatic inhibition activity (54.12–86.80%) than the ethanolic extract (23.07–81.61%).
In China, raspberry leaves are consumed as tea, and it has been reported to have anti-diabetic activity [112]. In a recent study, Li et al. investigated the inhibitory activity of raspberry leaf tea (Rubus corchorifolius L.) (RLT) extract digestive enzymes and found potent α-amylase inhibition activity for its ethanolic, methanolic, and aqueous extracts with an IC50 of 1.26 mg/mL, 1.47 mg/mL, and 4.39 mg/mL, respectively, against an IC50 of 5.12 mg/mL for acarbose aqueous extract [102]. The study identified the major inhibitors responsible for the extract activity as epigallocatechin gallate (EGCG), isovitexin, rutin, isoorientin, procyanidin, delphinidin-3-O-glucoside, dihydromyricetin, and procyanidin C3. The authors conducted additional molecular docking analyses and discovered these inhibitors interact with the digestive enzyme through hydrogen bonds or van der Waals forces, resulting in the retardation of enzyme activity [102]. Mikailu et al. studied the effect of the stem bark Maesobotrya dusenii Hutch methanol extract on α-amylase inhibition activity. The authors found that all the extracts showed significant amylase activity with 56.7% inhibition in a dose-dependent manner. Maesobotrya dusenii Hutch crude methanol extract showed α-amylase activity with an IC50 of 24 μg/mL against 28 μg/mL for acarbose.
Sterculia nobilis Smith seeds, leaves, and nuts are used to prepare several food dishes in China. It is commonly native to Vietnam, Indonesia, Japan, and south China and is usually utilized to heal gastrointestinal and circulatory problems [113,114,115]. The dark red shell of S. nobilis Smith fruit (pericarp) has been investigated for its inhibition activity against digestive enzymes [103]. The study showed that the pericarp ethyl acetate fraction has a potent uncompetitive α-amylase activity, with an IC50 of 13.55 μg/mL against 19.45 μg/mL for acarbose. Spectroscopic methods were used to elaborate the mechanism of α-amylase inhibition, and the results showed the ethyl acetate fraction alters the enzyme’s secondary structure and tryptophan/tyrosine residue microenvironment, resulting in enzyme activity inhibition [103]. The early investigation of these traditional medicinal plant extracts demonstrated promising α-amylase inhibitory activity, but more research is needed to confirm their anti-diabetic impact.
Prosopis cineraria (L.) Druce pods are usually used in diets as a vegetable in the Indian subcontinent and have been reported to have anti-diabetic properties [116]. Kumar et al. studied the anti-diabetic effect of P. cineraria pod extracts in vitro as well as in vivo (Table 2) [78]. The study shows that n-butanol fractions from the pods have potent α-amylase inhibition activity with an IC50 of 22.01 μg/mL against 39.26 μg/mL for acarbose. The n-butanol fraction was investigated for toxicity and found to be non-toxic when the mice ingested an oral dose of the fraction up to 2000 mg/kg. Further studies are required to investigate the P. cineraria pods as a promising anti-diabetic candidate. A recent study showed Terfeziaclaveryi (truffle, a fungus that grows wildly in the desert) methanol extract to have an α-amylase inhibition activity of 38.7 μg/mL, which is higher when compared to 45.3 μg/mL of acarbose [80]. The in vivo anti-diabetic activity showed that a 200 mg/kg dose of Terfeziaclaveryi methanol extract reduced the fasting plasma glucose level. Chenopodium album L. is another anti-diabetic herbal remedy candidate that showed promising α-amylase activity from its methanolic and aqueous root extracts [117,118]. C. album aerial parts’ alkaloid fraction (CAAF) showed a more potent α-amylase enzyme inhibition activity than acarbose, with an IC50 of 122.18 μg/mL and 812.83 μg/mL, respectively [91]. The CAAF fraction did not produce severe toxicity in vivo and showed promising anti-diabetic activity in a dose-dependent manner. The study suggests that the CAAF acts primarily as an α-amylase inhibitor. Salvia lavandulifolia Vahl is commonly found in the Mediterranean basin, and as a traditional medicine, it is applied as a virucidal, fungicidal, and bactericidal agent [119,120]. Its aqueous extract is widely used in this region as an anti-diabetic remedy for its hypoglycemic effect [121]. Remo et al. investigated the aqueous extract of S. lavandulifolia Vahl for its α-amylase inhibitory activity as well as its hypoglycemic effect in vivo [96]. The aqueous extract showed significant α-amylase inhibition activity with an IC50 of 0.99 mg/mL compared to an IC50 of 0.52 mg/mL for acarbose. It also showed a hypoglycemic effect on diabetic rats, with an AUC of 51.94 g/L/h. This study shows that S. lavandulifolia is a potential candidate for anti-diabetic drugs (Table 2).

4. Secondary Metabolites Isolated from Various Plant Sources as Potential α-Amylase Inhibitors

Many different plant extracts have been used for therapeutic purposes throughout history, whether on purpose or by accident. There have been reports of numerous plant extracts and their secondary metabolites having anti-diabetic effects, particularly through α-amylase or α-glucosidase inhibition [130,131]. Due to their fewer adverse side effects and easy accessibility, there is a growing interest in developing medicinal plant extracts or the extracted secondary metabolites as an alternative and complementary natural therapeutic for the medical care of diabetes [132,133]. Several secondary metabolite groups, including flavonoids, polysaccharides, phenolic acids, terpenoids, tannins, alkaloids, and xanthones, have been identified as prospective inhibitors of the α-amylase enzyme (Figure 4, Table 3).

4.1. Flavonoids

Flavonoids are the most ubiquitous class of secondary metabolites present in plants [164]. They are broadly studied due to their wide range of bioactivities, which include anti-oxidant [77], anti-inflammatory [165], anti-microbial [166], and anti-diabetic properties [167]. Numerous groups of flavonoids have the potential to inhibit α-amylase enzymes due to their non-covalent binding ability to the active site residues of the enzyme [168].
A flavone derivative, 5-hydroxy-2-(4-methoxy-3-((E)-3-methylbut-1-enyl)-5-(3-methylbut-3-enyl)phenyl)chroman-4-one (Figure 5a), identified in Andrographis echioides leaf, was found to dramatically inhibit α-amylase (IC50 = 3.35 µg/mL) and enhance glucose intake in the 3T3-L1 and L6 cell lines [134]. Another study conducted by Zhang et al. [136] revealed that epicatechin gallate (Figure 5b) (IC50 = 0.92 mg/mL) isolated from Euryale ferox seed coat possessed good inhibitory effects against α-amylase in comparison to acarbose (IC50 = 1.08 mg/mL). Similarly, Wu and Tian [138] isolated a new flavone glycoside, tricetin 4′-O-β-glucopyranoside (Figure 5c), along with a known flavone, Tricetin (Figure 5d), from the flowers of Punica granatum. Tricetin 4′-O-β-glucopyranoside (IC50 = 1.17 mg/mL) and Tricetin (IC50 = 0.43 mg/mL) both exhibited α-amylase inhibitory activities comparable to acarbose (IC50 = 0.03mg/mL). In addition, a new α-glucosidase and α-amylase inhibitor flavonoid named hypolaetin 8-O-β-D-galactopyranoside (Figure 5e) was isolated from the leaves of Thymelaea tartonraira [140].
Yang et al. [135] separated luteolin (Figure 5f) from Taraxacum mongolicum, which inhibits the α-amylase enzyme (IC50 = 42.33 µg/mL). From the molecular docking studies, they found that luteolin restricts the bioactivity of α-amylase by making a stable complex with the enzyme through Van der Waals forces, hydrogen bonding, and hydrophobic interaction. In a recent study, Mohamed and his co-workers [139] assessed 12 different flavonoids from Tagetes minuta for α-amylase inhibitory activities by in vitro experiments and in silico analyses. Compared to acarbose, quercetagetin-7-O-β-D-glucopyranoside (Figure 5g) showed the best in vitro enzyme inhibition (IC50 of 7.8 µM), more stability, and the highest binding affinity with the receptor in in silico studies. Such encouraging results are prompting us to further explore the flavonoid molecules and their interaction with α-amylase enzymes in in vivo studies, toxicity assessments, and the development of new anti-diabetic therapeutics.

4.2. Terpenoids

Terpenoids are among the diverse classes of natural compounds with potent medicinal qualities such as anti-cancer, anti-inflammatory, and antiviral properties [169]. They are major constituents of essential oils produced by aromatic plants and contribute to their flavor and fragrance [170]. Medicinal plant-derived terpenoids have been found to have promising hypoglycemic effects [171].
As part of the search for new anti-diabetic compounds from plants, Luyen et al. [141] examined the Vietnamese medicinal plant Wedelia trilobata, which was well-known for its efficacy in treating type 2 diabetes. They discovered wedtrilosides A and B (Figure 6a,b), two new ent-kaurane diterpenoids that have α-amylase inhibition activities. Furthermore, abietane diterpenes, carnosol (Figure 6c) and 12-methoxycarnosic acid (Figure 6d), isolated from Salvia aurita, revealed strong α-amylase inhibition with an IC50 of 19.8 and 16.2 µg/mL, respectively [142]. Similarly, oleanolic acid (Figure 6e) from Salvia Africana-lutea exhibited an α-amylase inhibition property with an IC50 of 12.5 µg/mL, close to acarbose (IC50 = 10.2 µg/mL) [172]. Another new terpenoid saponin molecule, ligularoside A (Figure 6f), was reported for the first time from Passiflora ligularis Juss leaves and showed a comparable inhibitory effect over α-amylase with an IC50 of 409 μM in an in vitro assay against acarbose, which had an IC50 of 234 μM [145].
Verma et al. [144] evaluated the anti-diabetic property of a novel triterpenoid, glochidon (Figure 6g), isolated from Phyllanthus debilis by in vitro assays, in vivo trials, and computational studies. In the in vitro assays, glochidon showed excellent α-amylase inhibition (IC50 = 38.15 μM), which is almost the same as that of acarbose. Additionally, it showed dose-dependent hypoglycemic effects in STZ-induced diabetic rats. Furthermore, the molecular dynamic simulations demonstrated that glochidon has a higher propensity to interact with the GLUT1 receptor protein. These findings can be utilized in the development of terpenoid-based natural medicine for both the management and avoidance of diabetes and its intricacies.

4.3. Polysaccharides

Polysaccharides derived from plants are macromolecules that consist of the same or different monosaccharide units interconnected with α- or β-glycosidic linkages. They have lengthy chains composed of carbohydrate molecules arranged either linearly as in amylose and cellulose or branched as in amylopectin and glycogen [173,174]. The composition, structure, and molecular weight of the polysaccharides vary with the plant species. Polysaccharide composition, molecular weight, linkage types, and chain patterns all influence their physical properties, such as solubility and viscosity, as well as chemical properties affecting pharmacological effects [175,176]. Plant polysaccharides have received a lot of interest in recent years due to their noteworthy bioactivities, non-toxic nature, and suitability for use in medicine [177,178,179]. Recent studies showed that many plant polysaccharides inhibit the α-amylase enzyme and play a crucial role in the treatment of diabetes [180,181,182].
It has been identified that the polysaccharides from Lycium barbarum leaves hinder α-amylase enzyme activity in a dose-dependent manner [149]. Another polysaccharide (SGP-1-1), isolated from Siraitia grosvenorii, showed α-amylase inhibition with the best inhibition of 61.73% [148]. Similarly, Zeng et al. [128] exploited the red seaweed laver-derived polysaccharide as a hypoglycemic agent. The polysaccharide named PD-1 showed 98.78% inhibition effects on the α-amylase enzyme with an IC50 of 12.72 mg/mL. Furthermore, the kinetic studies reveal that PD-1 interacts with the α-amylase enzyme in a competitive manner. Another group of researchers purified two polysaccharides, WSRP-2a (MW 56.8 kD) and WSRP-2b (MW 23.9 kD), from Rosa Setate x Rosa Rugosa biomass waste. The inhibition of the α-amylase enzyme by WSRP-2b (IC50 = 1.72 mg/mL) was much stronger compared to WSRP-2a (IC50 = 3.41 mg/mL), which may be related to changes in molecular weight [147]. Jiang and his coworkers used the ultrasonic-assisted extraction method to extract tamarind (Xyloglucan). When compared to Xyloglucan extracted using the hot water approach, they discovered that the ultrasound-aided extraction method greatly improved its α-amylase inhibitory activity (72.49%) by successfully reducing the viscosity and molecular weight of the compound [150]. In light of these encouraging results, additional research and examinations are needed to determine the correlation between the molecular structure and the anti-diabetic properties of polysaccharides and to create anti-diabetic medications derived from plant polysaccharides.

4.4. Phenolic Acids

Phenolic acids are the derivates of cinnamic and benzoic acids and are found in wide varieties of plants, either in free or bound forms [183]. Phenolic acids have been known for diverse bioactivities, including anti-microbial, antioxidant, anti-diabetic, and anti-cancer activities [184]. In carbohydrate metabolism, phenolic acids have been best known for their ability to inhibit the bioactivity of the α-glucosidase and α-amylase enzymes [185].
Wu et al. [74] studied the α-amylase inhibitory effects of ellagic acid (Figure 7a) obtained from corn kernels, which showed better inhibition (IC50 = 0.19 mg/mL) than the control (IC50 = 0.24 mg/mL). Similarly, Paun et al. [76] isolated rosmarinic acid (Figure 7b) and chlorogenic acids (Figure 7c) from Anchusa officinalis. Among these phenolic acids, rosmarinic acid (IC50 = 0.92 μg/mL) showed almost 20 fold higher inhibitory effects on the α-amylase enzyme than the standard sample (IC50 = 17.68 μg/mL). Likewise, another phenolic acid, p-coumaric acid (Figure 7d), identified in Agave americana L., showed considerable inhibition of the human α-amylase enzyme with an IC50 of 10.16 μM, which was around 2.3 times higher than the control [137]. In addition, the kinetic studies revealed that p-coumaric acid functions as a competitive α-amylase enzyme inhibitor. To fully realize the potential of phenolic acid compounds as a novel therapeutic for treating diabetes, more study is required to evaluate their efficacy, bioavailability, and safety.

4.5. Tannins

Tannins are polyphenolic biomolecules that have the tendency to precipitate proteins in water. They are widely available in many plant species and act as growth regulators and protectors against plant predators [186]. In traditional folk medicine, a variety of tannin-rich plants are frequently used by diabetic patients to treat diabetes mellitus and its associated issues [187].
Wang et al. [153] studied the in vitro α-amylase inhibition of Chinese bayberry leaf-derived proanthocyanidins (BLPs) and found that BLPs could retard the activity of an α-amylase enzyme using a mixed-type inhibition mode with an IC50 of 3.07 μg/mL. Chen et al. [151] identified twenty-five major ellagitannins from the unripe fruit of Rubus chingii Hu. Among them, chingiitannin A (Figure 8a) demonstrated around seven times higher inhibition with an IC50 of 4.52 μM compared to acarbose (IC50 = 35.71 μM). Moreover, molecular docking studies revealed that chingiitannin A binds at the allosteric site and primarily bonds with the enzymes through hydrogen bonds. Furthermore, chingiitannin A was non-toxic and improved glucose absorption. Similarly, 1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose (Figure 8b), from the leaves of Rhus coriaria, was reported to exhibit higher inhibitory effects on an α-amylase enzyme with an IC50 of 6.32 μM than acarbose (IC50 = 10.69 μM) [70]. However, more scientific analysis is necessary to determine the in vivo anti-diabetic effect of these tannins, and an assessment of toxicity should be performed in order to design therapeutic biomolecules with no or very few side effects.

4.6. Miscellaneous Secondary Metabolites as α-Amylase Inhibitors

Plants possess various bioactive compounds with different structural moieties, which demonstrated efficient α-amylase inhibition. For example, Thuy and his co-workers isolated three new pregnane glycosides from Dregea volubilis leaves, among which Drevoluoside Q exhibited significant inhibitory effects against α-amylase (IC50 = 51.3 µM), which is comparable to acarbose (IC50 = 36.3 µM) [157]. Likewise, phytochemical studies of another plant species, Gymnema sylvestre, resulted in the separation of five new pregnane glycosides, known as gymsylodide A, gymsylodide B, gymsylodide C, gymsylodide D, and gymsylodide E. Among them, gymsylodides B-E were found to have considerable α-amylase inhibitory action, with half-maximal inhibitory concentrations ranging from 113.0 to 176.2 µM [158].
An alkaloid, 3,3′,5,5′,8-pentamethyl-3,3′-bis(4-methylpent-3-en-1-yl)-3,3′,11,11′-tetrahydro-10,10′-bipyrano[3,2-a]carbazole (Figure 9a), was identified from Murraya koenigii leaves, which displayed significant in vitro α-amylase inhibition with an IC50 of 30.32 ppm [155]. Similarly, another known alkaloid, berberine (Figure 9b), was isolated from the Cardiospermum halicacabum plant and found to retard the activity of α-amylase by 72% at 10 μg/mL. In addition, in silico investigations indicated that berberine adheres to the enzyme’s active site [156].
Another class of bioactive molecules, xanthones named garcixanthone D and garcinone E (Figure 9c), were purified from the pericarp of Garcinia mangostana and showed 93.8 and 85.6% inhibition against α-amylase, respectively. The molecular docking studies revealed that these two compounds interact with α-amylase differently than the standard acarbose [160]. Similarly, a new prenylated xanthone, mangoxanthone A, with a moderate α-amylase inhibitory effect (IC50 = 22.74 μM), was reported from Garcinia mangostana pericarp [159].
Makinde et al. [162] discovered a fatty acid derivative, 5,7-dihydroxy-6-oxoheptadecanoic acid (Figure 9d), from the Tiliacora triandra plant that inhibited α-amylase enzymes (IC50 = 26.27 μM) more effectively than acarbose (IC50 = 177.65 μM). Halfordin (Figure 9e), a coumarin molecule purified from the bark extract of Melicope latifolia, was investigated for its potential to inhibit α-amylase enzymes [71] and was shown to be effective (IC50 = 197.53 μM). In addition, in silico investigations revealed a large number of molecular interactions with key α-amylase amino acid residues. Moreover, β-Sitosterol (Figure 9f) was identified as an effective α-amylase inhibitor from Parthenium hysterophorus leaf extract [163]. According to the molecular dynamics analyses, β-sitosterol has a larger binding energy than acarbose and the highest stability with α-amylase enzymes.
Investigations into natural compounds with significant α-amylase inhibitory effects are critical since many phytocompounds are consumed in the form of food in our daily lives. Based on findings on their potential effectiveness in inhibiting α-amylase enzymes, bioactive natural compounds are thought to play a very critical role in diabetes treatment. As a result, more research and analyses are needed to assess the toxic effect as well as other drug interactions or unfavorable effects that they might create in in vivo studies. Furthermore, additional human clinical trials are required to validate the safety and anti-diabetic benefits of α-amylase inhibitors derived from various plant species.

5. Conclusions

Diabetes is a chronic metabolic illness recognized by persistently elevated blood sugar levels resulting from a decrease in insulin synthesis or a rise in insulin resistance. One preliminary-stage diabetes therapy technique is to lessen post-meal hyperglycemia. This can be accomplished by moderating the bioactivity of α-amylases, carbohydrate-digestive enzymes that hinder glucose absorption in the gut. Consequently, inhibitors of these enzymes limit the rate of glucose absorption, lowering postprandial plasma glucose levels. Growing evidence from traditional and herbal approaches to diabetes management indicates that plant extracts and their chemical components may play a substantial role in the therapeutic management of T2D and its implications. Hence, this review study aimed to summarize the latest and most advanced findings in natural product research that operate as α-amylase enzyme inhibitors. Despite the fact that scientists have put in a lot of effort to identify the inhibitory effects of various plant extracts and natural compounds against the α-amylase enzyme, there are still some gaps. The majority of research focuses on in vitro investigations of the inhibitory effect of plant extracts and the identification of bioactive molecules. However, only a very limited amount of additional study was conducted for in vivo tests in animals, and human trials were extremely rare. Therefore, more research and scientific analyses are needed to assimilate the pharmacological activity of natural extracts and compounds, the synergistic effects of active compounds with other molecules, and their safer use. Moreover, clinical trials are crucial for reaching precise conclusions on both the safety and effectiveness of administering plant extracts or active compounds for the management of type 2 diabetes. Prospective studies will offer important information for determining the doses of natural extracts and active compounds that will deliver the desired therapeutic advantages while exhibiting little to no adverse side effects. This study explored over fifty extracts obtained with various solvents and over sixty natural ingredients. The insights gained from this review’s findings should help to achieve the ultimate goal of developing new therapeutic drugs with improved effectiveness and safety for the management of T2D or to avoid PPHG.

Author Contributions

Conceptualization, H.K.; Writing—original draft preparation, H.K.; Writing—review and editing, K.-H.B.; Funding acquisition, K.-H.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education (NRF-2021R1F1A1060297).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kahn, S.E.; Cooper, M.E.; Del Prato, S. Pathophysiology and Treatment of Type 2 Diabetes: Perspectives on the Past, Present, and Future. Lancet 2014, 383, 1068–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kharroubi, A.T. Diabetes Mellitus: The Epidemic of the Century. World J. Diabetes 2015, 6, 850. [Google Scholar] [CrossRef] [PubMed]
  3. Eizirik, D.L.; Pasquali, L.; Cnop, M. Pancreatic β-Cells in Type 1 and Type 2 Diabetes Mellitus: Different Pathways to Failure. Nat. Rev. Endocrinol. 2020, 16, 349–362. [Google Scholar] [CrossRef]
  4. Pinhas-Hamiel, O.; Zeitler, P. Acute and Chronic Complications of Type 2 Diabetes Mellitus in Children and Adolescents. Lancet 2007, 369, 1823–1831. [Google Scholar] [CrossRef] [PubMed]
  5. Forbes, J.M.; Cooper, M.E. Mechanisms of Diabetic Complications. Physiol. Rev. 2013, 93, 137–188. [Google Scholar] [CrossRef]
  6. Association, A.D. Standards of Medical Care in Diabetes—2014. Diabetes Care 2014, 37, S14–S80. [Google Scholar] [CrossRef] [Green Version]
  7. American Diabetes Association Professional Practice Committee. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2022. Diabetes Care 2022, 45, S17–S38. [Google Scholar] [CrossRef]
  8. Danaei, G.; Finucane, M.M.; Lu, Y.; Singh, G.M.; Cowan, M.J.; Paciorek, C.J.; Lin, J.K.; Farzadfar, F.; Khang, Y.-H.; Stevens, G.A.; et al. National, Regional, and Global Trends in Fasting Plasma Glucose and Diabetes Prevalence since 1980: Systematic Analysis of Health Examination Surveys and Epidemiological Studies with 370 Country-Years and 2·7 Million Participants. Lancet 2011, 378, 31–40. [Google Scholar] [CrossRef]
  9. International Diabetes Federation. IDF Diabetes Atlas, 10th ed.; IDF: Brussels, Belgium, 2021; Available online: https://diabetesatlas.org/idfawp/resource-files/2021/07/idf_atlas_10th_edition_2021.pdf (accessed on 1 May 2023).
  10. Alberti, K.G.M.M.; Zimmet, P.Z. Definition, Diagnosis and Classification of Diabetes Mellitus and Its Complications. Part 1: Diagnosis and Classification of Diabetes Mellitus. Provisional Report of a WHO Consultation. Diabet. Med. 1998, 15, 539–553. [Google Scholar] [CrossRef]
  11. Holman, R.R.; Paul, S.K.; Bethel, M.A.; Matthews, D.R.; Neil, H.A.W. 10-Year Follow-up of Intensive Glucose Control in Type 2 Diabetes. N. Engl. J. Med. 2008, 359, 1577–1589. [Google Scholar] [CrossRef] [Green Version]
  12. Papoutsis, K.; Zhang, J.; Bowyer, M.C.; Brunton, N.; Gibney, E.R.; Lyng, J. Fruit, Vegetables, and Mushrooms for the Preparation of Extracts with α-Amylase and α-Glucosidase Inhibition Properties: A Review. Food Chem. 2021, 338, 128119. [Google Scholar] [CrossRef]
  13. Tundis, R.; Loizzo, M.R.M.R.; Menichini, F. Natural Products as α-Amylase and & α-Glucosidase Inhibitors and Their Hypoglycaemic Potential in the Treatment of Diabetes: An Update. Mini-Rev. Med. Chem. 2010, 10, 315–331. [Google Scholar] [CrossRef] [PubMed]
  14. Watanabe, J.; Kawabata, J.; Kurihara, H.; Niki, R. Isolation and Identification of α -Glucosidase Inhibitors from Tochu-Cha (Eucommia Ulmoides). Biosci. Biotechnol. Biochem. 1997, 61, 177–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Horii, S.; Fukase, H.; Matsuo, T.; Kameda, Y.; Asano, N.; Matsui, K. Synthesis and. Alpha.-D-Glucosidase Inhibitory Activity of N-Substituted Valiolamine Derivatives as Potential Oral Antidiabetic Agents. J. Med. Chem. 1986, 29, 1038–1046. [Google Scholar] [CrossRef] [PubMed]
  16. Laar, F. Alpha-Glucosidase Inhibitors in the Early Treatment of Type 2 Diabetes. Vasc. Health Risk Manag. 2008, 4, 1189–1195. [Google Scholar] [CrossRef] [Green Version]
  17. Van de Laar, F.A.; Lucassen, P.L.; Akkermans, R.P.; Van de Lisdonk, E.H.; Rutten, G.E.; Van Weel, C. Alpha-Glucosidase Inhibitors for Type 2 Diabetes Mellitus. Cochrane Database Syst. Rev. 2005, 2009. [Google Scholar] [CrossRef] [Green Version]
  18. Ho, P.M.; Rumsfeld, J.S.; Masoudi, F.A.; McClure, D.L.; Plomondon, M.E.; Steiner, J.F.; Magid, D.J. Effect of Medication Nonadherence on Hospitalization and Mortality among Patients with Diabetes Mellitus. Arch. Intern. Med. 2006, 166, 1836. [Google Scholar] [CrossRef] [Green Version]
  19. Kashtoh, H.; Hussain, S.; Khan, A.; Saad, S.M.; Khan, J.A.J.; Khan, K.M.; Perveen, S.; Choudhary, M.I. Oxadiazoles and Thiadiazoles: Novel α-Glucosidase Inhibitors. Bioorganic Med. Chem. 2014, 22, 5454–5465. [Google Scholar] [CrossRef]
  20. Niaz, H.; Kashtoh, H.; Khan, J.A.J.; Khan, A.; Wahab, A.T.; Alam, M.T.; Khan, K.M.; Perveen, S.; Choudhary, M.I. Synthesis of Diethyl 4-Substituted-2,6-Dimethyl-1,4-Dihydropyridine-3,5-Dicarboxylates as a New Series of Inhibitors against Yeast α-Glucosidase. Eur. J. Med. Chem. 2015, 95, 199–209. [Google Scholar] [CrossRef]
  21. Kashtoh, H.; Muhammad, M.T.; Khan, J.J.A.; Rasheed, S.; Khan, A.; Perveen, S.; Javaid, K.; Atia-Tul-Wahab; Khan, K.M.; Choudhary, M.I. Dihydropyrano [2,3-c] Pyrazole: Novel in Vitro Inhibitors of Yeast α-Glucosidase. Bioorg. Chem. 2016, 65, 61–72. [Google Scholar] [CrossRef]
  22. Zhang, X.; World Health Organization. Traditional Medicine Strategy 2002–2005; World Health Organization: Geneva, Switzerland, 2002. [Google Scholar]
  23. Al-Aboudi, A.; Afifi, F.U. Plants Used for the Treatment of Diabetes in Jordan: A Review of Scientific Evidence. Pharm. Biol. 2011, 49, 221–239. [Google Scholar] [CrossRef]
  24. World Health Organization. WHO Expert Committee on Diabetes Mellitus [Meeting Held in Geneva from 25 September to 1 October 1979]: Second Report; World Health Organization: Geneva, Switzerland, 1980; ISBN 9241206462. [Google Scholar]
  25. Butterworth, P.J.; Warren, F.J.; Ellis, P.R. Human α-Amylase and Starch Digestion: An Interesting Marriage. Starch—Stärke 2011, 63, 395–405. [Google Scholar] [CrossRef]
  26. Patel, H.; Royall, P.G.; Gaisford, S.; Williams, G.R.; Edwards, C.H.; Warren, F.J.; Flanagan, B.M.; Ellis, P.R.; Butterworth, P.J. Structural and Enzyme Kinetic Studies of Retrograded Starch: Inhibition of α-Amylase and Consequences for Intestinal Digestion of Starch. Carbohydr. Polym. 2017, 164, 154–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Svensson, B. Regional Distant Sequence Homology between Amylases, α-Glucosidases and Transglucanosylases. FEBS Lett. 1988, 230, 72–76. [Google Scholar] [CrossRef] [Green Version]
  28. Henrissat, B. A Classification of Glycosyl Hydrolases Based on Amino Acid Sequence Similarities. Biochem. J. 1991, 280, 309–316. [Google Scholar] [CrossRef] [PubMed]
  29. Plaunt, A.J.; de Lourdes Betancourt-Mendiola, M.; Smith, B.D. Biomolecule Recognition Using Transition Metal Ions and Hydrogen Bonding. In Comprehensive Supramolecular Chemistry II; Elsevier: Amsterdam, The Netherlands, 2017; pp. 575–591. [Google Scholar] [CrossRef]
  30. Steer, M.L.; Levitzki, A. The Metal Specificity of Mammalian α-Amylase as Revealed by Enzyme Activity and Structural Probes. FEBS Lett. 1973, 31, 89–92. [Google Scholar] [CrossRef] [Green Version]
  31. Vallee, B.L.; Stein, E.A.; Sumerwell, W.N.; Fischer, E.H. Metal Content of α-Amylases of Various Origins. J. Biol. Chem. 1959, 234, 2901–2905. [Google Scholar] [CrossRef]
  32. Levitzki, A.; Steer, M.L. The Allosteric Activation of Mammalian Alpha-Amylase by Chloride. Eur. J. Biochem. 1974, 41, 171–180. [Google Scholar] [CrossRef]
  33. Brayer, G.D.; Luo, Y.; Withers, S.G. The Structure of Human Pancreatic α -Amylase at 1.8 Å Resolution and Comparisons with Related Enzymes. Protein Sci. 1995, 4, 1730–1742. [Google Scholar] [CrossRef]
  34. Gumucio, D.L.; Wiebauer, K.; Caldwell, R.M.; Samuelson, L.C.; Meisler, M.H. Concerted Evolution of Human Amylase Genes. Mol. Cell. Biol. 1988, 8, 1197–1205. [Google Scholar] [CrossRef] [PubMed]
  35. Sales, P.M.; Souza, P.M.; Simeoni, L.A.; Magalhães, P.O.; Silveira, D. α-Amylase Inhibitors: A Review of Raw Material and Isolated Compounds from Plant Source. J. Pharm. Pharm. Sci. 2012, 15, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Liu, D.; Gao, H.; Tang, W.; Nie, S. Plant Non-Starch Polysaccharides That Inhibit Key Enzymes Linked to Type 2 Diabetes Mellitus. Ann. N. Y. Acad. Sci. 2017, 1401, 28–36. [Google Scholar] [CrossRef] [PubMed]
  37. Martinez-Gonzalez, A.I.; Díaz-Sánchez, Á.G.; de la Rosa, L.A.; Vargas-Requena, C.L.; Bustos-Jaimes, I.; Alvarez-Parrilla, E. Polyphenolic Compounds and Digestive Enzymes: In Vitro Non-Covalent Interactions. Molecules 2017, 22, 669. [Google Scholar] [CrossRef] [Green Version]
  38. Brayer, G.D.; Sidhu, G.; Maurus, R.; Rydberg, E.H.; Braun, C.; Wang, Y.; Nguyen, N.T.; Overall, C.M.; Withers, S.G. Subsite Mapping of the Human Pancreatic α-Amylase Active Site through Structural, Kinetic, and Mutagenesis Techniques. Biochemistry 2000, 39, 4778–4791. [Google Scholar] [CrossRef]
  39. Dhital, S.; Warren, F.J.; Butterworth, P.J.; Ellis, P.R.; Gidley, M.J. Mechanisms of Starch Digestion by α -Amylase—Structural Basis for Kinetic Properties. Crit. Rev. Food Sci. Nutr. 2017, 57, 875–892. [Google Scholar] [CrossRef]
  40. Truscheit, E.; Frommer, W.; Junge, B.; Müller, L.; Schmidt, D.D.; Wingender, W. Chemistry and Biochemistry of Microbialα-Glucosidase Inhibitors. Angew. Chemie Int. Ed. Engl. 1981, 20, 744–761. [Google Scholar] [CrossRef]
  41. Nichols, B.L.; Avery, S.; Sen, P.; Swallow, D.M.; Hahn, D.; Sterchi, E. The Maltase-Glucoamylase Gene: Common Ancestry to Sucrase-Isomaltase with Complementary Starch Digestion Activities. Proc. Natl. Acad. Sci. USA 2003, 100, 1432–1437. [Google Scholar] [CrossRef]
  42. Williamson, G. Possible Effects of Dietary Polyphenols on Sugar Absorption and Digestion. Mol. Nutr. Food Res. 2013, 57, 48–57. [Google Scholar] [CrossRef]
  43. Ramasubbu, N.; Paloth, V.; Luo, Y.; Brayer, G.D.; Levine, M.J. Structure of Human Salivary α-Amylase at 1.6 Å Resolution: Implications for Its Role in the Oral Cavity. Acta Crystallogr. Sect. D Biol. Crystallogr. 1996, 52, 435–446. [Google Scholar] [CrossRef]
  44. Gilles, C.; Astier, J.; Marchis-Mouren, G.; Cambillau, C.; Payan, F. Crystal Structure of Pig Pancreatic Alpha-Amylase Isoenzyme II, in Complex with the Carbohydrate Inhibitor Acarbose. Eur. J. Biochem. 1996, 238, 561–569. [Google Scholar] [CrossRef] [PubMed]
  45. Lifshitz, R.; Levitzki, A. Identity and Properties of the Chloride Effector Binding Site in Hog Pancreatic α-Amylase. Biochemistry 1976, 15, 1987–1993. [Google Scholar] [CrossRef]
  46. MacGregor, E.A.; Janeček, Š.; Svensson, B. Relationship of Sequence and Structure to Specificity in the α-Amylase Family of Enzymes. Biochim. Biophys. Acta—Protein Struct. Mol. Enzymol. 2001, 1546, 1–20. [Google Scholar] [CrossRef]
  47. Schrödinger, L.L.C.; DeLano, W. Pymol. PyMOL Mol. Graph. Syst. Version 2020, 2. Available online: https://pymol.org/2/support.html? (accessed on 1 April 2022).
  48. Nahoum, V.; Roux, G.; Anton, V.; Rougé, P.; Puigserver, A.; Bischoff, H.; Henrissat, B.; Payan, F. Crystal Structures of Human Pancreatic α-Amylase in Complex with Carbohydrate and Proteinaceous Inhibitors. Biochem. J. 2000, 346, 201–208. [Google Scholar] [CrossRef]
  49. Aghajari, N.; Feller, G.; Gerday, C.; Haser, R. Structural Basis of α-Amylase Activation by Chloride. Protein Sci. 2002, 11, 1435–1441. [Google Scholar] [CrossRef]
  50. Qian, M.; Haser, R.; Buisson, G.; Duee, E.; Payan, F. The Active Center of a Mammalian .Alpha.-Amylase. Structure of the Complex of a Pancreatic. Alpha.-Amylase with a Carbohydrate Inhibitor Refined to 2.2-.ANG. Resolution. Biochemistry 1994, 33, 6284–6294. [Google Scholar] [CrossRef] [PubMed]
  51. Feller, G.; le Bussy, O.; Houssier, C.; Gerday, C. Structural and Functional Aspects of Chloride Binding to Alteromonas Haloplanctis α-Amylase. J. Biol. Chem. 1996, 271, 23836–23841. [Google Scholar] [CrossRef] [Green Version]
  52. Robyt, J.F. Starch: Structure, Properties, Chemistry, and Enzymology. In Glycoscience; Springer: Berlin/Heidelberg, Germany, 2008; pp. 1437–1472. [Google Scholar] [CrossRef]
  53. Robyt, J.F.; French, D. Multiple Attack Hypothesis of α-Amylase Action: Action of Porcine Pancreatic, Human Salivary, and Aspergillus Oryzae α-Amylases. Arch. Biochem. Biophys. 1967, 122, 8–16. [Google Scholar] [CrossRef] [PubMed]
  54. Seigner, C.; Prodanov, E.; Marchis-Mouren, G. The Determination of Subsite Binding Energies of Porcine Pancreatic α-Amylase by Comparing Hydrolytic Activity towards Substrates. Biochim. Biophys. Acta—Protein Struct. Mol. Enzymol. 1987, 913, 200–209. [Google Scholar] [CrossRef]
  55. Jarald, E.; Joshi, S.B.; Jain, D. Diabetes and Herbal Medicines. Pharmacol. Ther. 2008, 7, 97–106. [Google Scholar]
  56. Hamdan, I.; Afifi, F. Studies on the in Vitro and in Vivo Hypoglycemic Activities of Some Medicinal Plants Used in Treatment of Diabetes in Jordanian Traditional Medicine. J. Ethnopharmacol. 2004, 93, 117–121. [Google Scholar] [CrossRef] [PubMed]
  57. Tag, H.; Kalita, P.; Dwivedi, P.; Das, A.K.; Namsa, N.D. Herbal Medicines Used in the Treatment of Diabetes Mellitus in Arunachal Himalaya, Northeast, India. J. Ethnopharmacol. 2012, 141, 786–795. [Google Scholar] [CrossRef] [PubMed]
  58. Bailey, C.J.; Day, C. Traditional Plant Medicines as Treatments for Diabetes. Diabetes Care 1989, 12, 553–564. [Google Scholar] [CrossRef] [PubMed]
  59. Baharvand-Ahmadi, B.; Bahmani, M.; Tajeddini, P.; Naghdi, N.; Rafieian-Kopaei, M. An Ethno-Medicinal Study of Medicinal Plants Used for the Treatment of Diabetes. J. Nephropathol. 2015, 5, 44–50. [Google Scholar] [CrossRef]
  60. Giovannini, P.; Howes, M.-J.R.; Edwards, S.E. Medicinal Plants Used in the Traditional Management of Diabetes and Its Sequelae in Central America: A Review. J. Ethnopharmacol. 2016, 184, 58–71. [Google Scholar] [CrossRef] [Green Version]
  61. Moreira, D.D.L.; Teixeira, S.S.; Monteiro, M.H.D.; De-Oliveira, A.C.A.X.; Paumgartten, F.J.R. Traditional Use and Safety of Herbal Medicines. Rev. Bras. Farmacogn. 2014, 24, 248–257. [Google Scholar] [CrossRef]
  62. Ekor, M. The Growing Use of Herbal Medicines: Issues Relating to Adverse Reactions and Challenges in Monitoring Safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef] [Green Version]
  63. Daoudi, N.E.; Bouhrim, M.; Ouassou, H.; Legssyer, A.; Mekhfi, H.; Ziyyat, A.; Aziz, M.; Bnouham, M. Inhibitory Effect of Roasted/Unroasted Argania spinosa Seeds Oil on α- Glucosidase, α-Amylase and Intestinal Glucose Absorption Activities. S. Afr. J. Bot. 2020, 135, 413–420. [Google Scholar] [CrossRef]
  64. Quek, A.; Kassim, N.K.; Ismail, A.; Latif, M.A.M.; Shaari, K.; Tan, D.C.; Lim, P.C. Identification of Dipeptidyl Peptidase-4 and α-Amylase Inhibitors from Melicope glabra (Blume) TG Hartley (Rutaceae) Using Liquid Chromatography Tandem Mass Spectrometry, In Vitro and In Silico Methods. Molecules 2020, 26, 1. [Google Scholar] [CrossRef]
  65. Tekulu, G.H.; Araya, E.M.; Mengesha, H.G. In Vitro α-Amylase Inhibitory Effect of TLC Isolates of Aloe megalacantha Baker and Aloe monticola Reynolds. BMC Complement. Altern. Med. 2019, 19, 206. [Google Scholar] [CrossRef] [PubMed]
  66. Panigrahy, S.K.; Kumar, A.; Bhatt, R. Hedychium coronarium Rhizomes: Promising Antidiabetic and Natural Inhibitor of α-Amylase and α-Glucosidase. J. Diet. Suppl. 2020, 17, 81–87. [Google Scholar] [CrossRef] [PubMed]
  67. Hawash, M.; Jaradat, N.; Elaraj, J.; Hamdan, A.; Lebdeh, S.A.; Halawa, T. Evaluation of the Hypoglycemic Effect of Seven Wild Folkloric Edible Plants from Palestine (Antidiabetic Effect of Seven Plants from Palestine). J. Complement. Integr. Med. 2020, 17, 1–10. [Google Scholar] [CrossRef]
  68. Sansenya, S.; Payaka, A.; Wannasut, W.; Hua, Y.; Chumanee, S. Biological Activity of Rice Extract and the Inhibition Potential of Rice Extract, Rice Volatile Compounds and Their Combination against α-Glucosidase, α-Amylase and Tyrosinase. Int. J. Food Sci. Technol. 2021, 56, 1865–1876. [Google Scholar] [CrossRef]
  69. Xu, S.; Qin, L.; Mazhar, M.; Zhu, Y. Functional Components Profile and Glycemic Index of Kidney Beans. Front. Nutr. 2022, 9, 1044427. [Google Scholar] [CrossRef] [PubMed]
  70. Gök, H.N.; Deliorman Orhan, D.; Gürbüz, İ.; Aslan, M. Activity-Guided Isolation of α-Amylase, α-Glucosidase, and Pancreatic Lipase Inhibitory Compounds from Rhus coriaria L. J. Food Sci. 2020, 85, 3220–3228. [Google Scholar] [CrossRef] [PubMed]
  71. Quek, A.; Kassim, N.K.; Lim, P.C.; Tan, D.C.; Mohammad Latif, M.A.; Ismail, A.; Shaari, K.; Awang, K. α-Amylase and Dipeptidyl Peptidase-4 (DPP-4) Inhibitory Effects of Melicope latifolia Bark Extracts and Identification of Bioactive Constituents Using in Vitro and in Silico Approaches. Pharm. Biol. 2021, 59, 964–973. [Google Scholar] [CrossRef]
  72. Sani, D.H.; Munna, A.N.; Alam, M.J.; Salim, M.; Alam, M.J. Evaluation of α-Amylase Inhibition and Cytotoxic Activities of the Arachis hypogaea and Cinnamomum tamala. Curr. Nutr. Food Sci. 2020, 17, 328–336. [Google Scholar] [CrossRef]
  73. Romero Rocamora, C.; Ramasamy, K.; Meng Lim, S.; Majeed, A.B.A.; Agatonovic-Kustrin, S. HPTLC Based Approach for Bioassay-Guided Evaluation of Antidiabetic and Neuroprotective Effects of Eight Essential Oils of the Lamiaceae Family Plants. J. Pharm. Biomed. Anal. 2020, 178, 112909. [Google Scholar] [CrossRef]
  74. Wu, M.; Yang, Q.; Wu, Y.; Ouyang, J. Inhibitory Effects of Acorn (Quercus variabilis Blume) Kernel-Derived Polyphenols on the Activities of α-Amylase, α-Glucosidase, and Dipeptidyl Peptidase IV. Food Biosci. 2021, 43, 101224. [Google Scholar] [CrossRef]
  75. Sen, A.; Kurkcuoglu, M.; Yildirim, A.; Senkardes, I.; Bitis, L.; Baser, K.H.C. Chemical Composition, Antiradical, and Enzyme Inhibitory Potential of Essential Oil Obtained from Aerial Part of Centaurea pterocaula Trautv. J. Essent. Oil Res. 2021, 33, 44–52. [Google Scholar] [CrossRef]
  76. Paun, G.; Neagu, E.; Albu, C.; Savin, S.; Radu, G.L. In Vitro Evaluation of Antidiabetic and Anti-Inflammatory Activities of Polyphenolic-Rich Extracts from Anchusa officinalis and Melilotus officinalis. ACS Omega 2020, 5, 13014–13022. [Google Scholar] [CrossRef] [PubMed]
  77. Hoang Anh, L.; Xuan, T.D.; Dieu Thuy, N.T.; Van Quan, N.; Trang, L.T. Antioxidant and α-Amylase Inhibitory Activities and Phytocompounds of Clausena indica Fruits. Medicines 2020, 7, 10. [Google Scholar] [CrossRef] [Green Version]
  78. Kumar, M.; Govindasamy, J.; Nyola, N.K. In-Vitro and in-Vivo Anti-Hyperglycemic Potential of Prosopis cineraria Pods Extract and Fractions. J. Biol. Act. Prod. Nat. 2019, 9, 135–140. [Google Scholar] [CrossRef]
  79. Prasathkumar, M.; Raja, K.; Vasanth, K.; Khusro, A.; Sadhasivam, S.; Sahibzada, M.U.K.; Gawwad, M.R.A.; Al Farraj, D.A.; Elshikh, M.S. Phytochemical Screening and in Vitro Antibacterial, Antioxidant, Anti-Inflammatory, Anti-Diabetic, and Wound Healing Attributes of Senna auriculata (L.) Roxb. Leaves. Arab. J. Chem. 2021, 14, 103345. [Google Scholar] [CrossRef]
  80. Al-Ahmed, A.; Khalil, H.E. Antidiabetic Activity of Terfeziaclaveryi; An in Vitro and in Vivo Study. Biomed. Pharmacol. J. 2019, 12, 603–608. [Google Scholar] [CrossRef]
  81. Prakash, V. Determination of α-Amylase Inhibitory Potential of Leaf Extracts of Rhododendron arboreum Sm. and Rhododendron campanulatum D. Don. J. Drug Deliv. Ther. 2022, 12, 20–22. [Google Scholar] [CrossRef]
  82. Mazumder, K.; Sumi, T.S.; Golder, M.; Biswas, B.; Maknoon; Kerr, P.G. Antidiabetic Profiling, Cytotoxicity and Acute Toxicity Evaluation of Aerial Parts of Phragmites karka (Retz.). J. Ethnopharmacol. 2021, 270, 113781. [Google Scholar] [CrossRef]
  83. da Silva, D.H.A.; Barbosa, H.d.M.; Beltrão, R.L.d.A.; Silva, C.d.F.O.; Moura, C.A.; Castro, R.N.; Almeida, J.R.G.d.S.; Gomes, D.A.; Lira, E.C. Hexane Fraction from Brazilian Morus nigra Leaves Improved Oral Carbohydrate Tolerance and Inhibits α-Amylase and α-Glucosidase Activities in Diabetic Mice. Nat. Prod. Res. 2021, 35, 4785–4788. [Google Scholar] [CrossRef]
  84. Ktari, N.; Ben Slama-Ben Salem, R.; Bkhairia, I.; Ben Slima, S.; Nasri, R.; Ben Salah, R.; Nasri, M. Functional Properties and Biological Activities of Peptides from Zebra Blenny Protein Hydrolysates Fractionated Using Ultrafiltration. Food Biosci. 2020, 34, 100539. [Google Scholar] [CrossRef]
  85. Renganathan, S.; Manokaran, S.; Vasanthakumar, P.; Singaravelu, U.; Kim, P.S.; Kutzner, A.; Heese, K. Phytochemical Profiling in Conjunction with in Vitro and in Silico Studies to Identify Human α-Amylase Inhibitors in Leucaena leucocephala (Lam.) de Wit for the Treatment of Diabetes Mellitus. ACS Omega 2021, 6, 19045–19057. [Google Scholar] [CrossRef] [PubMed]
  86. Pandey, B.P.; Pradhan, S.P.; Adhikari, K.; Nepal, S. Bergenia pacumbis from Nepal, an Astonishing Enzymes Inhibitor. BMC Complement. Med. Ther. 2020, 20, 198. [Google Scholar] [CrossRef]
  87. Jaradat, N.; Abualhasan, M.N.; Qadi, M.; Issa, L.; Mousa, A.; Allan, F.; Hindi, M.; Alhrezat, Z. Antiamylase, Antilipase, Antimicrobial, and Cytotoxic Activity of Nonea obtusifolia (Willd.) Dc. From Palestine. Biomed Res. Int. 2020, 2020, 8821319. [Google Scholar] [CrossRef] [PubMed]
  88. Jan, B.; Zahiruddin, S.; Basist, P.; Irfan, M.; Abass, S.; Ahmad, S. Metabolomic Profiling and Identification of Antioxidant and Antidiabetic Compounds from Leaves of Different Varieties of Morus alba Linn Grown in Kashmir. ACS Omega 2022, 7, 24317–24328. [Google Scholar] [CrossRef]
  89. Omar, R.M.; Badria, F.A.; Galala, A.A. An Emerging Flavone Glycoside from Phyllanthus emblica L.: As Promiscuous Enzyme Inhibitor and Potential Therapeutic in Chronic Diseases. S. Afr. J. Bot. 2023, 153, 290–296. [Google Scholar] [CrossRef]
  90. Timalsina, D.; Bhusal, D.; Devkota, H.P.; Pokhrel, K.P.; Sharma, K.R. α -Amylase Inhibitory Activity of Catunaregam spinosa (Thunb.) Tirveng.: In Vitro and in Silico Studies. Biomed Res. Int. 2021, 2021, 4133876. [Google Scholar] [CrossRef]
  91. Choudhary, N.; Prabhakar, P.K.; Khatik, G.L.; Chamakuri, S.R.; Tewari, D.; Suttee, A. Evaluation of Acute Toxicity, in-Vitro, in-Vivo Antidiabetic Potential of the Flavonoid Fraction of the Plant Chenopodium album L. Pharmacogn. J. 2021, 13, 765–779. [Google Scholar] [CrossRef]
  92. Saraswathi, K.; Bharkavi, R.; Khusro, A.; Sivaraj, C.; Arumugam, P.; Alghamdi, S.; Dablool, A.S.; Almehmadi, M.; Bannunah, A.M.; Umar Khayam Sahibzada, M. Assessment on in Vitro Medicinal Properties and Chemical Composition Analysis of Solanum virginianum Dried Fruits. Arab. J. Chem. 2021, 14, 103442. [Google Scholar] [CrossRef]
  93. Bello, M.; Jiddah-kazeem, B.; Fatoki, T.H.; Ibukun, E.O.; Akinmoladun, A.C. Antioxidant Property of Eucalyptus globulus Labill. Extracts and Inhibitory Activities on Carbohydrate Metabolizing Enzymes Related to Type-2 Diabetes. Biocatal. Agric. Biotechnol. 2021, 36, 102111. [Google Scholar] [CrossRef]
  94. Mikailu, S.; Okorafor, M.C.; Abo, K.A. Alpha-Amylase Inhibition and Membrane Stabilizing Effect of the Stem Bark of Maesobotrya dusenii Hutchinson. Int. J. Pharm. Sci. Res. 2019, 10, 5154–5159. [Google Scholar] [CrossRef]
  95. Hassan, A.; Khan Mohmand, N.Z.; Ullah, H.; Hussain, A. Antioxidant, Antidiabetic, and Antihypertension Inhibitory Potentials of Phenolic Rich Medicinal Plants. J. Chem. 2022, 2022, 9046780. [Google Scholar] [CrossRef]
  96. Remok, F.; Saidi, S.; Gourich, A.A.; Zibouh, K.; Maouloua, M.; El Makhoukhi, F.; El Menyiy, N.; Touijer, H.; Bouhrim, M.; Sahpaz, S.; et al. Phenolic Content, Antioxidant, Antibacterial, Antihyperglycemic, and α-Amylase Inhibitory Activities of Aqueous Extract of Salvia lavandulifolia Vahl. Pharmaceuticals 2023, 16, 395. [Google Scholar] [CrossRef] [PubMed]
  97. Karray, A.; Alonazi, M.; Jallouli, R.; Alanazi, H.; Ben Bacha, A. A Proteinaceous Alpha-Amylase Inhibitor from Moringa oleifera Leaf Extract: Purification, Characterization, and Insecticide Effects against C. Maculates Insect Larvae. Molecules 2022, 27, 4222. [Google Scholar] [CrossRef] [PubMed]
  98. Ukwuani-Kwaja, A.; Sani, I.; Sylvester, M.N.N.; Kabir, A.O.; Abolaji, O.K.; Ukwuani-Kwaja, A.; Sani, I.; Sylvester, M.N.N. In-Vitro Antidiabetic Effect of Ziziphus mucronata Leave Extracts. J. Drug Deliv. Ther. 2021, 11, 9–13. [Google Scholar] [CrossRef]
  99. Olaokun, O.O.; Manonga, S.A.; Zubair, M.S.; Maulana, S.; Mkolo, N.M. Molecular Docking and Molecular Dynamics Studies of Antidiabetic Phenolic Compound Isolated from Leaf Extract of Englerophytum magalismontanum (Sond.) T.D.Penn. Molecules 2022, 27, 3175. [Google Scholar] [CrossRef] [PubMed]
  100. Yashoda, K.; Deegendra, K.; Bimala, S. Antioxidant, Ptp 1B Inhibition and A-Amylase Inhibition Property and Gc-Ms Analysis of Methanolic Leaves Extract of Achyranthes aspera and Catharanthus roseus of Nepal. Int. J. Pharm. Pharm. Sci. 2021, 13, 49–55. [Google Scholar] [CrossRef]
  101. Irfan Dar, M.; Qureshi, M.I.; Zahiruddin, S.; Abass, S.; Jan, B.; Sultan, A.; Ahmad, S. In Silico Analysis of PTP1B Inhibitors and TLC-MS Bioautography-Based Identification of Free Radical Scavenging and α-Amylase Inhibitory Compounds from Heartwood Extract of Pterocarpus marsupium. ACS Omega 2022, 7, 46156–46173. [Google Scholar] [CrossRef]
  102. Li, F.; Luo, T.; Hou, J.; Fei, T.; Zhang, J.; Wang, L. Natural α-Glucosidase and α-Amylase Inhibitors from Raspberry (Rubus corchorifolius L.) Leaf-Tea: Screening, Identification and Molecular Docking Analysis. Lwt 2023, 181, 114763. [Google Scholar] [CrossRef]
  103. Yang, Y.; Zhang, P.; Huang, Z.; Zhao, Z. Phenolics from Sterculia nobilis Smith Pericarp By-Products Delay Carbohydrate Digestion by Uncompetitively Inhibiting α-Glucosidase and α-Amylase. LWT 2023, 173, 114339. [Google Scholar] [CrossRef]
  104. Sakhr, K.; El Khatib, S. Physiochemical Properties and Medicinal, Nutritional and Industrial Applications of Lebanese Sumac (Syrian Sumac—Rhus coriaria): A Review. Heliyon 2020, 6, e03207. [Google Scholar] [CrossRef] [Green Version]
  105. Pleşca-Manea, L.; Pârvu, A.E.; Pârvu, M.; Taămaş, M.; Buia, R.; Puia, M. Effects of Melilotus officinalis on Acute Inflammation. Phyther. Res. 2002, 16, 316–319. [Google Scholar] [CrossRef] [PubMed]
  106. Burlando, B.; Verotta, L.; Cornara, L.; Bottini-Massa, E. Herbal Principles in Cosmetics: Properties and Mechanisms of Action; CRC Press: Boca Raton, FL, USA, 2010; ISBN 1439812144. [Google Scholar]
  107. Bakhshayeshi, S.; Madani, S.; Hemmatabadi, M.; Heshmat, R.; Larijani, B. Effects of Semelil (ANGIPARSTM) on Diabetic Peripheral Neuropathy: A Randomized, Double-Blind Placebo-Controlled Clinical Trial. Daru 2011, 19, 65–70. [Google Scholar]
  108. Nithya, M.; Ragavendran, C.; Natarajan, D. Antibacterial and Free Radical Scavenging Activity of a Medicinal Plant Solanum Xanthocarpum. Int. J. Food Prop. 2018, 21, 313–327. [Google Scholar] [CrossRef] [Green Version]
  109. Tekuri, S.K.; Pasupuleti, S.K.; Konidala, K.K.; Amuru, S.R.; Bassaiahgari, P.; Pabbaraju, N. Phytochemical and Pharmacological Activities of Solanum Surattense Burm. f.–A Review. J. Appl. Pharm. Sci. 2019, 9, 126–136. [Google Scholar] [CrossRef] [Green Version]
  110. Pereira, A.P.A.; Lauretti, L.B.C.; Alvarenga, V.O.; Paulino, B.N.; Angolini, C.F.F.; Neri-Numa, I.A.; Orlando, E.A.; Pallone, J.A.L.; Sant’Ana, A.S.; Pastore, G.M. Evaluation of Fruta-Do-Lobo (Solanum Lycocarpum St. Hill) Starch on the Growth of Probiotic Strains. Food Res. Int. 2020, 133, 109187. [Google Scholar] [CrossRef] [PubMed]
  111. Pereira, A.P.A.; Fernando Figueiredo Angolini, C.; de Souza-Sporkens, J.C.; da Silva, T.A.; Coutinho Franco de Oliveira, H.; Pastore, G.M. Brazilian Sunberry (Solanum Oocarpum Sendtn): Alkaloid Composition and Improvement of Mitochondrial Functionality and Insulin Secretion of INS-1E Cells. Food Res. Int. 2021, 148, 110589. [Google Scholar] [CrossRef]
  112. Wu, L.; Liu, Y.; Qin, Y.; Wang, L.; Wu, Z. HPLC-ESI-QTOF-MS/MS Characterization, Antioxidant Activities and Inhibitory Ability of Digestive Enzymes with Molecular Docking Analysis of Various Parts of Raspberry (Rubus ideaus L.). Antioxidants 2019, 8, 274. [Google Scholar] [CrossRef] [Green Version]
  113. Song, J.; Sun, P.; Wang, R.; Zhao, X. Gastroprotective Effects of Methanolic Extract of S Terculia Nobilis Smith Seeds in Reserpine-Induced Gastric Ulcer in Mice. J. Food Biochem. 2015, 39, 230–237. [Google Scholar] [CrossRef]
  114. Zhang, Y.; Shen, R.; Mo, Y.; Li, Q.; Lin, W.; Yuan, G. Colletotrichum Siamense: A Novel Leaf Pathogen of Sterculia nobilis Smith Detected in China. For. Pathol. 2020, 50, e12575. [Google Scholar] [CrossRef]
  115. Zhang, J.-J.; Li, Y.; Lin, S.-J.; Li, H.-B. Green Extraction of Natural Antioxidants from the Sterculia nobilis Fruit Waste and Analysis of Phenolic Profile. Molecules 2018, 23, 1059. [Google Scholar] [CrossRef] [Green Version]
  116. Sharma, N.; Garg, V.; Paul, A. Antihyperglycemic, Antihyperlipidemic and Antioxidative Potential of Prosopis cineraria Bark. Indian J. Clin. Biochem. 2010, 25, 193–200. [Google Scholar] [CrossRef] [Green Version]
  117. Kant, S. Pharmacological Evaluation of Antidiabetic and Antihyperlipidemic Activity of Chenopodium album Root Extract in Male Wistar Albino Rat Models. Int. J. Green Pharm. 2018, 12, 115–122. [Google Scholar] [CrossRef]
  118. Odhav, B.; Kandasamy, T.; Khumalo, N.; Baijnath, H. Screening of African Traditional Vegetables for Their Alpha-Amylase Inhibitory Effect. J. Med. Plants Res 2010, 4, 1502–1507. [Google Scholar] [CrossRef]
  119. Jordán, M.J.; Martínez, C.; Moñino, M.I.; Lax, V.; Quílez, M.; Sotomayor, J.A. Chemical Characterization of Salvia lavandulifolia Subsp. Vellerea in South-Eastern Spain. Acta Hortic. 2009, 317–324. [Google Scholar] [CrossRef]
  120. Zrira, S.; Menut, C.; Bessiere, J.M.; Elamrani, A.; Benjilali, B. A Study of the Essential Oil of Salvia lavandulifolia Vahl from Morocco. J. Essent. Oil Bear. Plants 2004, 7, 232–238. [Google Scholar] [CrossRef]
  121. Zarzuelo, A.; Risco, S.; Gámez, M.J.; Jimenez, J.; Cámara, M.; Martinez, M.A. Hypoglycemic Action of Vahl. ssp.: A Contribution to Studies of the Mechanism of Action. Life Sci. 1990, 47, 909–915. [Google Scholar] [CrossRef]
  122. Kabach, I.; Bouchmaa, N.; Zouaoui, Z.; Ennoury, A.; El Asri, S.; Laabar, A.; Oumeslakht, L.; Cacciola, F.; El Majdoub, Y.O.; Mondello, L.; et al. Phytochemical Profile and Antioxidant Capacity, α-Amylase and α-Glucosidase Inhibitory Activities of Oxalis pes-caprae Extracts in Alloxan-Induced Diabetic Mice. Biomed. Pharmacother. 2023, 160, 114393. [Google Scholar] [CrossRef] [PubMed]
  123. Malik, A.; Sharif, A.; Zubair, H.M.; Akhtar, B.; Mobashar, A. In Vitro, In Silico, and In Vivo Studies of Cardamine hirsuta Linn as a Potential Antidiabetic Agent in a Rat Model. ACS Omega 2023, 8, 22623–22636. [Google Scholar] [CrossRef]
  124. Wan, P.; Cai, B.; Chen, H.; Chen, D.; Zhao, X.; Yuan, H.; Huang, J.; Chen, X.; Luo, L.; Pan, J. Antidiabetic Effects of Protein Hydrolysates from Trachinotus ovatus and Identification and Screening of Peptides with α-Amylase and DPP-IV Inhibitory Activities. Curr. Res. Food Sci. 2023, 6, 100446. [Google Scholar] [CrossRef] [PubMed]
  125. Naz, F.; Zahoor, M.; Ayaz, M.; Ashraf, M.; Nawaz, A.; Alotaibi, A. Anti-Diabetic Potentials of Sorbaria tomentosa Lindl. Rehder: Phytochemistry (GC-MS Analysis), α-Amylase, α-Glucosidase Inhibitory, in Vivo Hypoglycemic, and Biochemical Analysis. Open Chem. 2023, 21, 20220339. [Google Scholar] [CrossRef]
  126. Hbika, A.; Daoudi, N.E.; Bouyanzer, A.; Bouhrim, M.; Mohti, H.; Loukili, E.H.; Mechchate, H.; Al-Salahi, R.; Nasr, F.A.; Bnouham, M.; et al. Artemisia absinthium L. Aqueous and Ethyl Acetate Extracts: Antioxidant Effect and Potential Activity In Vitro and In Vivo against Pancreatic α-Amylase and Intestinal α-Glucosidase. Pharmaceutics 2022, 14, 481. [Google Scholar] [CrossRef]
  127. Bouknana, S.; Daoudi, N.E.; Bouhrim, M.; Ziyyat, A.; Legssyer, A.; Mekhfi, H.; Bnouham, M. Ammodaucus leucotrichus Coss. & Durieu: Antihyperglycemic Activity via the Inhibition of α-Amylase, α-Glucosidase, and Intestinal Glucose Absorption Activities and Its Chemical Composition. J. Pharm. Pharmacogn. Res. 2022, 10, 94–103. [Google Scholar] [CrossRef]
  128. Zeng, A.; Yang, R.; Yu, S.; Zhao, W. A Novel Hypoglycemic Agent: Polysaccharides from Laver (Porphyra spp.). Food Funct. 2020, 11, 9048–9056. [Google Scholar] [CrossRef] [PubMed]
  129. Xie, C.; Gao, W.; Li, X.; Luo, S.; Wu, D.; Chye, F.Y. Garlic (Allium sativum L.) Polysaccharide Ameliorates Type 2 Diabetes Mellitus (T2DM) via the Regulation of Hepatic Glycogen Metabolism. NFS J. 2023, 31, 19–27. [Google Scholar] [CrossRef]
  130. Kashtoh, H.; Baek, K.-H. Recent Updates on Phytoconstituent Alpha-Glucosidase Inhibitors: An Approach towards the Treatment of Type Two Diabetes. Plants 2022, 11, 2722. [Google Scholar] [CrossRef] [PubMed]
  131. Lam, T.; Tran, N.N.; Pham, L.D.; Lai, N.V.; Ngoc, B. Flavonoids as Dual- Target Inhibitors against α—Glucosidase and α -Amylase: A Systematic Review of in Vitro Studies. Biol. Med. Chem. 2023, 1–41. [Google Scholar] [CrossRef]
  132. Etxeberria, U.; De La Garza, A.L.; Campin, J.; Martnez, J.A.; Milagro, F.I. Antidiabetic Effects of Natural Plant Extracts via Inhibition of Carbohydrate Hydrolysis Enzymes with Emphasis on Pancreatic Alpha Amylase. Expert Opin. Ther. Targets 2012, 16, 269–297. [Google Scholar] [CrossRef] [Green Version]
  133. Gong, L.; Feng, D.; Wang, T.; Ren, Y.; Liu, Y.; Wang, J. Inhibitors of α-Amylase and α-Glucosidase: Potential Linkage for Whole Cereal Foods on Prevention of Hyperglycemia. Food Sci. Nutr. 2020, 8, 6320–6337. [Google Scholar] [CrossRef]
  134. Sivalingam, S.; Kandhasamy, S.; Chandrasekaran, S.; Vijayan, K.; Jacob, J.P.; Perumal, A.; Vijayakumar, S. A Flavone Derivative from Andrographis Echioides Leaf Extract Positively Alters the Molecular Targets of Insulin Signaling Pathway. S. Afr. J. Bot. 2022, 146, 760–770. [Google Scholar] [CrossRef]
  135. Yang, Y.; Wang, Y.; Zeng, W.; Tian, J.; Zhao, X.; Han, J.; Huang, D.; Gu, D. A Strategy Based on Liquid-Liquid-Refining Extraction and High-Speed Counter-Current Chromatography for the Bioassay-Guided Separation of Active Compound from Taraxacum Mongolicum. J. Chromatogr. A 2020, 1614, 460727. [Google Scholar] [CrossRef]
  136. Zhang, L.; Zeng, J.; Yuan, E.; Chen, J.; Zhang, Q.; Wang, Z.; Yin, Z. Extraction, Identification, and Starch-Digestion Inhibition of Phenolics from Euryale Ferox Seed Coat. J. Sci. Food Agric. 2023, 103, 3437–3446. [Google Scholar] [CrossRef]
  137. Sahnoun, M.; Bejar, S.; Daoud, L.; Ayadi, L.; Brini, F.; Saibi, W. Effect of Agave americana L. on the Human, and Aspergillus Oryzae S2 α-Amylase Inhibitions. Nat. Prod. Res. 2019, 33, 755–758. [Google Scholar] [CrossRef] [PubMed]
  138. Wu, S.; Tian, L. A New Flavone Glucoside Together with Known Ellagitannins and Flavones with Anti-Diabetic and Anti-Obesity Activities from the Flowers of Pomegranate (Punica granatum). Nat. Prod. Res. 2019, 33, 252–257. [Google Scholar] [CrossRef] [PubMed]
  139. Mohamed, G.A.; Omar, A.M.; El-Araby, M.E.; Mass, S.; Ibrahim, S.R.M. Assessments of Alpha-Amylase Inhibitory Potential of Tagetes Flavonoids through In Vitro, Molecular Docking, and Molecular Dynamics Simulation Studies. Int. J. Mol. Sci. 2023, 24, 10195. [Google Scholar] [CrossRef] [PubMed]
  140. Soltani, S.; Koubaa, I.; Dhouib, I.; Khemakhem, B.; Marchand, P.; Allouche, N. New Specific A—Glucosidase Inhibitor Flavonoid from Thymelaea Tartonraira Leaves: Structure Elucidation, Biological and Molecular Docking Studies. Chem. Biodivers. 2023, 20, e202200944. [Google Scholar] [CrossRef]
  141. Luyen, N.T.; Binh, P.T.; Tham, P.T.; Hung, T.M.; Dang, N.H.; Dat, N.T.; Thao, N.P. Wedtrilosides A and B, Two New Diterpenoid Glycosides from the Leaves of Wedelia trilobata (L.) Hitchc. with α-Amylase and α-Glucosidase Inhibitory Activities. Bioorg. Chem. 2019, 85, 319–324. [Google Scholar] [CrossRef]
  142. Etsassala, N.G.E.R.; Badmus, J.A.; Marnewick, J.L.; Iwuoha, E.I.; Nchu, F.; Hussein, A.A. Alpha-Glucosidase and Alpha-Amylase Inhibitory Activities, Molecular Docking, and Antioxidant Capacities of Salvia Aurita Constituents. Antioxidants 2020, 9, 1149. [Google Scholar] [CrossRef]
  143. Mohammed, A.; Victoria Awolola, G.; Ibrahim, M.A.; Anthony Koorbanally, N.; Islam, M.S. Oleanolic Acid as a Potential Antidiabetic Component of Xylopia Aethiopica (Dunal) A. Rich. (Annonaceae) Fruit: Bioassay Guided Isolation and Molecular Docking Studies. Nat. Prod. Res. 2021, 35, 788–791. [Google Scholar] [CrossRef]
  144. Verma, A.; Pathak, P.; Rimac, H.; Khalilullah, H.; Kumar, V.; Grishina, M.; Potemkin, V.; Ahmed, B. A Triterpene Glochidon from Phyllanthus Debilis: Isolation, Computational Studies, and Antidiabetic Activity Evaluation. Biocatal. Agric. Biotechnol. 2021, 36, 102138. [Google Scholar] [CrossRef]
  145. Monzón Daza, G.; Meneses Macías, C.; Forero, A.M.; Rodríguez, J.; Aragón, M.; Jiménez, C.; Ramos, F.A.; Castellanos, L. Identification of α-Amylase and α-Glucosidase Inhibitors and Ligularoside A, a New Triterpenoid Saponin from Passiflora Ligularis Juss (Sweet granadilla) Leaves, by a Nuclear Magnetic Resonance-Based Metabolomic Study. J. Agric. Food Chem. 2021, 69, 2919–2931. [Google Scholar] [CrossRef]
  146. Uddin, M.J.; Russo, D.; Haque, M.A.; Çiçek, S.S.; Sönnichsen, F.D.; Milella, L.; Zidorn, C. Bioactive Abietane-Type Diterpenoid Glycosides from Leaves of Clerodendrum infortunatum (Lamiaceae). Molecules 2021, 26, 4121. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, M.; Li, W.; Zhang, Y.; Shi, L.; Xu, Z.; Xia, W.; Zhang, W. Structure Characteristics, Hypoglycemic and Immunomodulatory Activities of Pectic Polysaccharides from Rosa Setate x Rosa Rugosa Waste. Carbohydr. Polym. 2021, 253, 117190. [Google Scholar] [CrossRef] [PubMed]
  148. Gong, P.; Guo, Y.; Chen, X.; Cui, D.; Wang, M.; Yang, W.; Chen, F. Structural Characteristics, Antioxidant and Hypoglycemic Activities of Polysaccharide from Siraitia Grosvenorii. Molecules 2022, 27, 4192. [Google Scholar] [CrossRef]
  149. Quan, N.; Wang, Y.D.; Li, G.R.; Liu, Z.Q.; Feng, J.; Qiao, C.L.; Zhang, H.F. Ultrasound–Microwave Combined Extraction of Novel Polysaccharide Fractions from Lycium Barbarum Leaves and Their In Vitro Hypoglycemic and Antioxidant Activities. Molecules 2023, 28, 3880. [Google Scholar] [CrossRef]
  150. Jiang, X.; Yang, T.; Li, Y.; Liu, S.; Liu, Y.; Chen, D.; Qin, W.; Zhang, Q.; Lin, D.; Liu, Y.; et al. Ultrasound-Assisted Extraction of Tamarind Xyloglucan: An Effective Approach to Reduce the Viscosity and Improve the α-Amylase Inhibition of Xyloglucan. J. Sci. Food Agric. 2022, 103, 4047–4057. [Google Scholar] [CrossRef]
  151. Chen, Y.; Chen, Z.; Guo, Q.; Gao, X.; Ma, Q.; Xue, Z.; Ferri, N.; Zhang, M.; Chen, H. Identification of Ellagitannins in the Unripe Fruit of Rubus Chingii Hu and Evaluation of Its Potential Antidiabetic Activity. J. Agric. Food Chem. 2019, 67, 7025–7039. [Google Scholar] [CrossRef]
  152. Cuc, N.T.; Cuong, N.T.; Anh, L.T.; Yen, D.T.H.; Tai, B.H.; Thu Trang, D.; Yen, P.H.; Van Kiem, P.; Nam, N.H.; Van Minh, C.; et al. Dihydrostilbene Glycosides from Camellia Sasanqua and Their α-Glucosidase and α-Amylase Inhibitory Activities. Nat. Prod. Res. 2021, 35, 4025–4031. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, M.; Chen, J.; Ye, X.; Liu, D. In Vitro Inhibitory Effects of Chinese Bayberry (Myrica rubra Sieb. et Zucc.) Leaves Proanthocyanidins on Pancreatic α-Amylase and Their Interaction. Bioorg. Chem. 2020, 101, 104029. [Google Scholar] [CrossRef]
  154. Dandekar, P.D.; Kotmale, A.S.; Chavan, S.R.; Kadlag, P.P.; Sawant, S.V.; Dhavale, D.D.; Ravikumar, A. Insights into the Inhibition Mechanism of Human Pancreatic α-Amylase, a Type 2 Diabetes Target, by Dehydrodieugenol B Isolated from Ocimum Tenuiflorum. ACS Omega 2021, 6, 1780–1786. [Google Scholar] [CrossRef] [PubMed]
  155. Sampath, S.N.T.I.; Jayasinghe, S.; Attanayake, A.P.; Karunaratne, V.; Yaddehige, M.L.; Watkins, D.L. A New Dimeric Carbazole Alkaloid from Murraya koenigii (L.) Leaves with α-Amylase and α-Glucosidase Inhibitory Activities. Phytochem. Lett. 2022, 52, 87–91. [Google Scholar] [CrossRef]
  156. Naik, S.; Deora, N.; Pal, S.K.; Ahmed, M.Z.; Alqahtani, A.S.; Shukla, P.K.; Venkatraman, K.; Kumar, S. Purification, Biochemical Characterization, and DPP-IV and α-Amylase Inhibitory Activity of Berberine from Cardiospermum Halicacabum. J. Mol. Recognit. 2022, 35, e2983. [Google Scholar] [CrossRef]
  157. Thuy, N.T.K.; Phuong, P.T.; Hien, N.T.T.; Trang, D.T.; Van Huan, N.; Anh, P.T.L.; Tai, B.H.; Nhiem, N.X.; Hung, N.T.; Kiem, P. Van Pregnane Glycosides from the Leaves of Dregea Volubilis and Their α-Glucosidase and α-Amylase Inhibitory Activities. Nat. Prod. Res. 2021, 35, 3931–3938. [Google Scholar] [CrossRef] [PubMed]
  158. Van Kiem, P.; Yen, D.T.H.; Van Hung, N.; Nhiem, N.X.; Tai, B.H.; Trang, D.T.; Yen, P.H.; Ngoc, T.M.; Van Minh, C.; Park, S.; et al. Five New Pregnane Glycosides from Gymnema Sylvestre and Their α-Glucosidase and α-Amylase Inhibitory Activities. Molecules 2020, 25, 2525. [Google Scholar] [CrossRef] [PubMed]
  159. Yang, L.; Zhang, D.; Li, J.-B.; Zhang, X.; Zhou, N.; Zhang, W.-Y.; Lu, H. Prenylated Xanthones with α-Glucosidase and α-Amylase Inhibitory Effects from the Pericarp of Garcinia Mangostana. J. Asian Nat. Prod. Res. 2022, 24, 624–633. [Google Scholar] [CrossRef]
  160. Ibrahim, S.R.M.; Mohamed, G.A.; Khayat, M.T.A.; Ahmed, S.; Abo-Haded, H. Garcixanthone D, a New Xanthone, and Other Xanthone Derivatives from Garcinia Mangostana Pericarps: Their α-Amylase Inhibitory Potential and Molecular Docking Studies. Starch/Staerke 2019, 71, 1800354. [Google Scholar] [CrossRef]
  161. 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]
  162. Makinde, E.A.; Ovatlarnporn, C.; Sontimuang, C.; Herbette, G.; Olatunji, O.J. Chemical Constituents from the Aerial Part of Tiliacora Triandra (Colebr.) Diels and Their α-Glucosidase and α-Amylase Inhibitory Activity. Nat. Prod. Commun. 2020, 15. [Google Scholar] [CrossRef]
  163. Ravi, L.; Girish, S.; D’Souza, S.R.; Anirudh Sreenivas, B.K.; Shree Kumari, G.R.; Archana, O.; Ajith Kumar, K.; Manjunathan, R. β-Sitosterol, a Phytocompound from Parthenium hysterophorus, Reveals Anti-Diabetic Properties through α-Amylase Inhibition: An i n-Silico and in-Vitro Analysis. J. Biomol. Struct. Dyn. 2023, 1–12. [Google Scholar] [CrossRef]
  164. Zhu, J.; Chen, C.; Zhang, B.; Huang, Q. The Inhibitory Effects of Flavonoids on α-Amylase and α-Glucosidase. Crit. Rev. Food Sci. Nutr. 2020, 60, 695–708. [Google Scholar] [CrossRef]
  165. Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-Inflammatory Effects of Flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef] [PubMed]
  166. Górniak, I.; Bartoszewski, R.; Króliczewski, J. Comprehensive Review of Antimicrobial Activities of Plant Flavonoids. Phytochem. Rev. 2019, 18, 241–272. [Google Scholar] [CrossRef] [Green Version]
  167. Proença, C.; Ribeiro, D.; Freitas, M.; Fernandes, E. Flavonoids as Potential Agents in the Management of Type 2 Diabetes through the Modulation of α-Amylase and α-Glucosidase Activity: A Review. Crit. Rev. Food Sci. Nutr. 2022, 62, 3137–3207. [Google Scholar] [CrossRef]
  168. Williams, L.K.; Li, C.; Withers, S.G.; Brayer, G.D. Order and Disorder: Differential Structural Impacts of Myricetin and Ethyl Caffeate on Human Amylase, an Antidiabetic Target. J. Med. Chem. 2012, 55, 10177–10186. [Google Scholar] [CrossRef]
  169. Ajikumar, P.K.; Tyo, K.; Carlsen, S.; Mucha, O.; Phon, T.H.; Stephanopoulos, G. Terpenoids: Opportunities for Biosynthesis of Natural Product Drugs Using Engineered Microorganisms. Mol. Pharm. 2008, 5, 167–190. [Google Scholar] [CrossRef] [PubMed]
  170. Masyita, A.; Mustika Sari, R.; Dwi Astuti, A.; Yasir, B.; Rahma Rumata, N.; Emran, T.B.; Nainu, F.; Simal-Gandara, J. Terpenes and Terpenoids as Main Bioactive Compounds of Essential Oils, Their Roles in Human Health and Potential Application as Natural Food Preservatives. Food Chem. X 2022, 13, 100217. [Google Scholar] [CrossRef] [PubMed]
  171. Teoh, S.L.; Das, S. Phytochemicals and Their Effective Role in the Treatment of Diabetes Mellitus: A Short Review. Phytochem. Rev. 2018, 17, 1111–1128. [Google Scholar] [CrossRef]
  172. Etsassala, N.G.E.R.; Badmus, J.A.; Waryo, T.T.; Marnewick, J.L.; Cupido, C.N.; Hussein, A.A.; Iwuoha, E.I. Alpha-Glucosidase and Alpha-Amylase Inhibitory Activities of Novel Abietane Diterpenes from Salvia Africana-Lutea. Antioxidants 2019, 8, 421. [Google Scholar] [CrossRef] [Green Version]
  173. He, L.Y.; Li, Y.; Niu, S.Q.; Bai, J.; Liu, S.J.; Guo, J.L. Polysaccharides from Natural Resource: Ameliorate Type 2 Diabetes Mellitus via Regulation of Oxidative Stress Network. Front. Pharmacol. 2023, 14, 1184572. [Google Scholar] [CrossRef]
  174. Wang, P.-C.; Zhao, S.; Yang, B.-Y.; Wang, Q.-H.; Kuang, H.-X. Anti-Diabetic Polysaccharides from Natural Sources: A Review. Carbohydr. Polym. 2016, 148, 86–97. [Google Scholar] [CrossRef] [PubMed]
  175. Ganesan, K.; Xu, B. Anti-Diabetic Effects and Mechanisms of Dietary Polysaccharides. Molecules 2019, 24, 2556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Wu, J.; Shi, S.; Wang, H.; Wang, S. Mechanisms Underlying the Effect of Polysaccharides in the Treatment of Type 2 Diabetes: A Review. Carbohydr. Polym. 2016, 144, 474–494. [Google Scholar] [CrossRef] [PubMed]
  177. Xiao, M.; Jia, X.; Wang, N.; Kang, J.; Hu, X.; Goff, H.D.; Cui, S.W.; Ding, H.; Guo, Q. Therapeutic Potential of Non-Starch Polysaccharides on Type 2 Diabetes: From Hypoglycemic Mechanism to Clinical Trials. Crit. Rev. Food Sci. Nutr. 2022, 1–34. [Google Scholar] [CrossRef]
  178. Tanna, B.; Mishra, A. Nutraceutical Potential of Seaweed Polysaccharides: Structure, Bioactivity, Safety, and Toxicity. Compr. Rev. Food Sci. Food Saf. 2019, 18, 817–831. [Google Scholar] [CrossRef]
  179. Sheng, K.; Wang, C.; Chen, B.; Kang, M.; Wang, M.; Liu, K.; Wang, M. Recent Advances in Polysaccharides from Lentinus Edodes (Berk.): Isolation, Structures and Bioactivities. Food Chem. 2021, 358, 129883. [Google Scholar] [CrossRef]
  180. Cao, C.; Li, C.; Chen, Q.; Huang, Q.; Pérez, M.E.M.; Fu, X. Physicochemical Characterization, Potential Antioxidant and Hypoglycemic Activity of Polysaccharide from Sargassum Pallidum. Int. J. Biol. Macromol. 2019, 139, 1009–1017. [Google Scholar] [CrossRef] [PubMed]
  181. Tan, M.; Zhao, Q.; Zhao, B. Physicochemical Properties, Structural Characterization and Biological Activities of Polysaccharides from Quinoa (Chenopodium quinoa Willd.) Seeds. Int. J. Biol. Macromol. 2021, 193, 1635–1644. [Google Scholar] [CrossRef]
  182. Li, Y.M.; Zhong, R.F.; Chen, J.; Luo, Z.G. Structural Characterization, Anticancer, Hypoglycemia and Immune Activities of Polysaccharides from Russula Virescens. Int. J. Biol. Macromol. 2021, 184, 380–392. [Google Scholar] [CrossRef]
  183. Bodoira, R.; Maestri, D. Phenolic Compounds from Nuts: Extraction, Chemical Profiles, and Bioactivity. J. Agric. Food Chem. 2020, 68, 927–942. [Google Scholar] [CrossRef] [PubMed]
  184. Kumar, N.; Goel, N. Phenolic Acids: Natural Versatile Molecules with Promising Therapeutic Applications. Biotechnol. Reports 2019, 24, e00370. [Google Scholar] [CrossRef]
  185. Vinayagam, R.; Jayachandran, M.; Xu, B. Antidiabetic Effects of Simple Phenolic Acids: A Comprehensive Review. Phyther. Res. 2016, 30, 184–199. [Google Scholar] [CrossRef]
  186. Marsh, K.J.; Wallis, I.R.; Kulheim, C.; Clark, R.; Nicolle, D.; Foley, W.J.; Salminen, J.P. New Approaches to Tannin Analysis of Leaves Can Be Used to Explain in Vitro Biological Activities Associated with Herbivore Defence. New Phytol. 2020, 225, 488–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ajebli, M.; Eddouks, M. The Promising Role of Plant Tannins as Bioactive Antidiabetic Agents. Curr. Med. Chem. 2018, 26, 4852–4884. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic depiction of the involvement of α-amylase in postprandial hyperglycemia linked with starch hydrolysis.
Figure 1. Schematic depiction of the involvement of α-amylase in postprandial hyperglycemia linked with starch hydrolysis.
Plants 12 02944 g001
Figure 2. (a) Ribbon diagram of the structure of human pancreatic α-amylase as a representative for GH13 α-amylase. The three structural domains (A, B, and C) are indicated and colored as follows: Domain A in blues, greens, yellows, and oranges; Domain B in lime green and pale cyan; and Domain C in red. The N and C terminals are colored blue and red, respectively. The calcium and chloride ions are shown as magenta and cyan spheres, respectively. (b) Human pancreatic α-amylase calcium binding site; the sticks represent residues making ligand interactions with calcium, which is represented as a magenta sphere. (c) Human pancreatic α-amylase chloride binding site; the sticks represent residues making ligand interactions with chloride, which is represented as a cyan sphere. From the structure, (ac) were adopted with PDB entry code 1HNY [33] and produced using PyMol [47].
Figure 2. (a) Ribbon diagram of the structure of human pancreatic α-amylase as a representative for GH13 α-amylase. The three structural domains (A, B, and C) are indicated and colored as follows: Domain A in blues, greens, yellows, and oranges; Domain B in lime green and pale cyan; and Domain C in red. The N and C terminals are colored blue and red, respectively. The calcium and chloride ions are shown as magenta and cyan spheres, respectively. (b) Human pancreatic α-amylase calcium binding site; the sticks represent residues making ligand interactions with calcium, which is represented as a magenta sphere. (c) Human pancreatic α-amylase chloride binding site; the sticks represent residues making ligand interactions with chloride, which is represented as a cyan sphere. From the structure, (ac) were adopted with PDB entry code 1HNY [33] and produced using PyMol [47].
Plants 12 02944 g002
Figure 3. (a) The structure of the human pancreatic α-amylase/acarbose complex is depicted as a ribbon diagram. The chloride and calcium ions are represented as orange and magenta spheres, respectively, in (a,b). (b) The active site of the human pancreatic α-amylase/acarbose complex; residues located within a 4-A° radius of acarbose are represented as sticks (the catalytic triad of D197-D300-E233). The acarbose, shown as sticks in (a,b), is colored cyan. From the structure, (a,b) were adopted with PDB entry code 1B2Y [48] and produced using PyMol [47].
Figure 3. (a) The structure of the human pancreatic α-amylase/acarbose complex is depicted as a ribbon diagram. The chloride and calcium ions are represented as orange and magenta spheres, respectively, in (a,b). (b) The active site of the human pancreatic α-amylase/acarbose complex; residues located within a 4-A° radius of acarbose are represented as sticks (the catalytic triad of D197-D300-E233). The acarbose, shown as sticks in (a,b), is colored cyan. From the structure, (a,b) were adopted with PDB entry code 1B2Y [48] and produced using PyMol [47].
Plants 12 02944 g003
Figure 4. A diagram depicting the numerous plant-derived compounds that are effective α-amylase inhibitors.
Figure 4. A diagram depicting the numerous plant-derived compounds that are effective α-amylase inhibitors.
Plants 12 02944 g004
Figure 5. Molecular structures of various plant-derived flavonoids identified as α-amylase inhibitors.
Figure 5. Molecular structures of various plant-derived flavonoids identified as α-amylase inhibitors.
Plants 12 02944 g005
Figure 6. Molecular structures of various plant-derived terpenoids identified as α-amylase inhibitors.
Figure 6. Molecular structures of various plant-derived terpenoids identified as α-amylase inhibitors.
Plants 12 02944 g006
Figure 7. Molecular structures of various plant-derived phenolic acids identified as α-amylase inhibitors.
Figure 7. Molecular structures of various plant-derived phenolic acids identified as α-amylase inhibitors.
Plants 12 02944 g007
Figure 8. Molecular structures of various plant-derived tannins identified as α-amylase inhibitors.
Figure 8. Molecular structures of various plant-derived tannins identified as α-amylase inhibitors.
Plants 12 02944 g008
Figure 9. Molecular structures of various plant-derived secondary metabolites identified as α-amylase inhibitors.
Figure 9. Molecular structures of various plant-derived secondary metabolites identified as α-amylase inhibitors.
Plants 12 02944 g009
Table 1. In Vitro α-amylase inhibition properties reported from various plant extracts.
Table 1. In Vitro α-amylase inhibition properties reported from various plant extracts.
Name of PlantsUsed Extract/Fraction/PartsIC50IC50 (Control)
(Acarbose)
Ref.
Argania spinosaRoasted seeds oil2.17 ± 0.24 mg/mL0.41 ± 0.015 mg/mL[63]
Unroasted seeds oil0.78 ± 0.16 mg/mL
Melicope glabra (Blume) T. G. HartleyChloroform extracts
(leaves)
303.64 ± 10.10 μg/mL188.6 ± 14.31 μg/mL[64]
Aloe megalacantha BakerLeaf latex74.76 ± 1.98 μg/mL16.49 ± 1.91 μg/mL[65]
Aloe monticola Reynolds 78.10 ± 1.88 μg/mL16.49 ± 1.91 μg/mL
Hedychium coronarium KoenEthyl acetate fraction
(rhizomes)
58.15 ± 1.23 μg/mL-[66]
Cichorium endiviaHydrophilic fraction (extracted from leaves with ethanol/water)9.96 μg/mL10.00 μg/mL[67]
Brown riceBrown rice (BR) extract48.96 ± 0.34%54.14 ± 0.35%[68]
BR/Vanillin99.32 ± 1.18%86.48 ± 0.71%
BR/Vanillyl alcohol96.55 ± 0.12%
Biyun no.7
(Kidney bean)
Aqueous extract1.659 ± 0.050 U/g DW-[69]
Rhus coriaria L.Ethyl acetate sub-extract
(leaves)
20.810 ± 0.747 μg/mL26.993 ± 0.797 μg/mL[70]
Melicope latifoliaChloroform extract
(bark)
1464.32 μg/mL [71]
Arachis hypogaeaEthanol extract
(peanut seeds)
0.61 μg/mL0.32 μg/mL[72]
Backhousia citriodoraEssential oil0.49 mg/μL-[73]
Rosmarinus officinalisEssential oil of rosemary plus0.45 mg/μL
Mentha piperitaEssential oil
(leaf)
0.41 mg/μL
Origanum vulgareEssential oil
(leaf-phenol type)
0.41 mg/μL
Quercus variabilis BlumeFree polyphenol extract5.25 ± 0.57 mg/mL0.24 mg/mL[74]
Bound polyphenol extract1.37 ± 0.11 mg/mL
Centaurea pterocaula TrautvEssential oil
(aerial part)
79.66 ± 0.43 µg/mL11.6 ± 0.18 µg/mL[75]
Anchusa officinalisCrude extract954.16 ± 7.46 µg/mL17.68 ± 1.24 µg/mL[76]
Melilotus officinalisCrude extract1.32 ± 0.08 µg/mL
Clausena indicaHexane extract1.37 ± 0.01 mg/mL0.07 ± 0.00 mg/mL[77]
Ethyl acetate extract8.56 ± 0.24 mg/mL
Prosopis cineraria (L.)n-Butanol fraction
(pods)
22.01 ± 0.92 μg/mL39.26 ± 2.19 μg/mL[78]
Ethyl acetate fraction28.23 ± 1.06 μg/mL
Senna auriculata (L.) Roxb.Methanolic extract
(leaves)
49.45 μg/mL-[79]
Terfezia claveryiMethanol extract38.7 μg/mL45.3 μg/mL[80]
Rhododendron arboreum Sm.Methanol extract51.1%-[81]
Phragmites karka (Retz.)Dichloromethane fraction
(aerial part)
2.05 mg/mL-
-
[82]
n-Hexane fraction
(aerial part)
2.08 mg/mL
Morus nigraHexane fraction
(leaves)
13.05 mg/mL0 0.21 mg/mL[83]
Salaria basiliscaProtein hydrolysates (peptide fraction F1)71 μg/mL14 μg/mL[84]
Leucaena leucocephala (Lam.) De WitEthanol extract
(leaves)
288.01 μg/mL252.59 μg/mL[85]
Bergenia pacumbisMethanol extract14.03 ± 0.04 μg/mL20.12 ± 0.12 μg/mL[86]
Nonea obtusifolia (Wild.) DC.Acetone extract25.7 ± 0.08 μg/mL28.18 ± 1.22 μg/mL[87]
Morus alba LinnLeaves74.76 ± 6.76 μg/mL35.34 ± 4.87 μg/mL[88]
Phylanthus emblica L.Methanolic extract
(leaves)
98.37 ± 1.09% [89]
Catunaregam spinosaDichloromethane fraction
(bark)
77.17 ± 1.75 μg/mL6.34 ± 0.07 μg/mL[90]
Chenopodium album L.Flavonoid fraction
(aerial part)
122.18 ± 1.15 μg/mL812.83 ± 1.07 μg/mL[91]
Solanum virginianumAqueous extract
(fruits)
54.12 ± 0.44–86.80 ± 0.27%58.36 ± 0.30–88.24 ± 0.16%[92]
Ethanolic extract
(fruits)
23.07 ± 0.47–81.61 ± 0.43%
Eucalyptus globulusEthanol extract (Hexane defatted)23.6 ± 1.2 μg/mL5.2 ± 1.3 μg/mL[93]
Ethanol extract (non-defatted)14.8 ± 1.2 μg/mL
Maesobotrya dusenii Hutch.Crude methanol extract24 μg/mL28 μg/mL[94]
Veronica bilobaAqueous extract110.25 μg/mL138.79 μg/mL[95]
Ethyl acetate extract121.09 μg/mL
Dichloromethane extract123.68 μg/mL
Salvia lavandulifolia VahlAqueous extract0.99 ± 0.00 mg/mL0.52 ± 0.01 mg/mL[96]
Moringa oleiferaMethanolic crude extract
(leaves)
65.6 ± 4.93%-[97]
Ziziphus mucronataAcetone extract0.62 mg/mL0.42 mg/mL[98]
Englerophytum magalismontanumMethanol fraction
(leaves)
10.76 ± 1.33 µg/mL1.24 ± 1.64 µg/mL[99]
Achyranthes asperaCrude extract97.60 ± 1.11 μg/mL68.13 ± 0.46 μg/mL[100]
Catharanthus roseus94.05 ± 1.18 μg/mL
Pterocarpus marsupiumMethanolic extract158.663 ± 10.986 μg/mL56.060 ± 4.465 μg/mL[101]
Rubus corchorifolius L.70% ethanolic extract
(leaf tea)
1.26 ± 0.03 mg/mL5.12 ± 0.42 mg/mL[102]
70% methanolic extract
(leaf tea)
1.47 ± 0.05 mg/mL
Aqueous extract
(leaf tea)
4.39 ± 0.17 mg/mL
Sterculia nobilis SmithEthyl acetate fraction
(pericarp)
13.550 ± 0.230 μg/mL19.45 ± 0.26 μg/mL[103]
Table 2. List of in vivo effects of α-amylase inhibitors reported from several plant extracts.
Table 2. List of in vivo effects of α-amylase inhibitors reported from several plant extracts.
Plant ExtractsDosage of Plant Extract UsedExperimental AnimalsTypes of Diabetes InductionAdministration RouteDiagnostic CriteriaInferenceRef.
Oxalis pes-caprae
Methanolic extract
150 mg/kg BWSwiss albino miceAlloxan-induced diabetesIntraperitoneal injectionFasting blood glucose (FBG) level, body weightHypoglycemic[122]
Cardamine hirsuta Linn
Hydro-methanolic extract
125, 250, and 500 mg/kg BWMale Sprague Dawley (SD) ratsHigh-fat diet (HFD), Streptozotocin (STZ)-induced diabetesOral and intraperitoneal injectionFBG levelHypoglycemic, dose-dependent[123]
Trachinotus ovatus
Protein hydrolysates
100, 500, and 1000 mg/kg BWMale Kunming miceSTZ-induced diabetesIntraperitoneal injectionFBG levelHypoglycemic, dose-dependent[124]
Sorbaria tomentosa Lindl. Rehder
Methanolic extract
150 and 300 mg/kg BWRatsAlloxan-induced diabetesIntraperitoneal injectionFBG levelHypoglycemic[125]
Chenopodium album L.
Flavonoid fraction
500 mg/kg BWSD ratsHFD-STZ-induced diabetesOral and intraperitoneal injectionGlucose, cholesterol, and triglyceride levelsHypoglycemic[91]
Terfezia claveryi Methanolic extract200 mg/kg BWMale Wistar albino ratsSTZ-induced diabetesIntraperitoneal injectionFBG levelHypoglycemic
time-dependent
[80]
Salvia lavandulifolia Vahl
Aqueous extract
400 mg/kg BWNormal ratsD-glucose, 2 g/kgOralOral glucose tolerance test (OGTT)
Blood glucose level
Hypoglycemic[96]
Artemisia absinthium L.
Aqueous extract
200 mg/kg BWWistar ratsAlloxan-induced diabetesOralPostprandial blood glucose (PBG) levelHypoglycemic[126]
Ammodaucus leucotrichus Coss. and Durieu
Aqueous extract
150 mg/kg BWWistar albino ratsAlloxan-induced diabetesOralOGTTHypoglycemic[127]
Porphyra spp. Polysaccharides100 mg/kg BWRats-OralPBG levelHypoglycemic[128]
Prosopis cineraria
Ethyl acetate fraction/n-Butanol fraction
250, 500, 1000 and 2000 mg/kg BWSwiss albino miceSucrose tolerance test (OSTT)OralSerum glucose concentrationHypoglycemic[78]
Allium sativum L.
Polysaccharides
1.25, 2.5, and 5 g/kg BWMale Kunming miceSTZ-induced diabetesIntraperitoneal injectionOGTT, FBGHypoglycemic[129]
Table 3. List of α-amylase inhibitors (in vitro) isolated from diverse plant species.
Table 3. List of α-amylase inhibitors (in vitro) isolated from diverse plant species.
Class of CompoundsBioactive CompoundSource (Plant’s Name)IC50IC50
(Control)
(Acarbose)
Ref.
Flavonoids5-hydroxy-2-(4-methoxy-3-((E)-3-methylbut-1-enyl)-5-(3-methylbut-3-enyl)phenyl)chroman-4-oneAndrographis echioides3.357 µg/mL [134]
LuteolinTaraxacum mongolicum42.33 ± 0.82 μg/mL-[135]
IsoquercitrinMelilotus officinalis9.65 ± 0.43 μg/mL17.68 ± 1.24 μg/mL[76]
Epicatechin gallateEuryale ferox
(seed coat)
0.92 mg/mL1.08 mg/mL[136]
PuerarinAgave americana L.3.87 μM [137]
TricetinPunica granatum0.43 ± 0.12 mg/mL0.038 ± 0.017 mg/mL[138]
Flavonoid glucosideTricetin 4′-O-β-glucopyranoside1.17 ± 0.32 mg/mL
RutinMelilotus officinalis11.42 ± 0.62 μg/mL17.68 ± 1.24 μg/mL[76]
quercetagetin-7-O-β-D-glucopyranosideTagetes minuta L.7.8 µM7.1 µM[139]
luteolin-7-O-α-L-rhamnosidePhylanthus emblica L.89.79%-[89]
hypolaetin 8-O-β-D-galactopyranosideThymelaea tartonraira
(leaves)
46.49 ± 2.32 μg/mL0.44 ± 0.022 μg/mL[140]
Ent-kaurane diterpenoidsWedtriloside AWedelia trilobata
(leaves)
112.20 ± 2.87 μg/mL-[141]
Wedtriloside B87.10 ± 1.89 μg/mL
Abietane diterpeneCarnosolSalvia aurita
(aerial part)
19.8 ± 1.4 µg/mL10.2 ± 0.6 µg/mL[142]
12-methoxycarnosic acid16.2 ± 0.3 µg/mL
Pentacyclic triterpenoidOleanolic acidXylopia aethiopica (Dunal) A. Rich.
(fruit)
89.02 ± 1.12 µM-[143]
TriterpenoidGlochidonPhyllanthus debilis38.15 ± 1.40 μM33.68 ± 3.12 μM[144]
Triterpenoid saponinLigularoside APassiflora ligularis Juss
(leaves)
409.8 ± 11.4 μM234.1 ± 15.9 μM[145]
PhenylpropanoidsJionoside DClerodendrum infortunatum L. 3.4 ± 0.2 μM5.9 ± 0.1 μM[146]
PolysaccharidesWSRP-2aRosa setate × Rosa rugosa
(waste biomass)
3.41 mg/mL0.57 mg/mL[147]
WSRP-2b1.72 mg/mL
PD-1Porphyra spp.12.72 mg/mL [128]
SGP-1-1Siraitia grosvenorii61.73% at 1 mg/mL [148]
LLP50 (polysaccharides fraction)Lycium barbarum
(leaves)
1.659 mg/mL0.0002 mg/mL[149]
XyloglucanTamarindus indica L72.49 ± 0.84%92.49 ± 1.97%[150]
Phenolic acid and its derivativesEllagic acidQuercus variabilis Blume0.19 ± 0.02 μg/mL0.24 μg/mL[74]
Rosmaric acidAnchusa officinalis0.92 ± 0.07 μg/mL17.68 ± 1.24 μg/mL[76]
Chlorogenic acid1.84 ± 0.05 μg/mL
p-Coumaric acidAgave americana L.10.16 μM-[137]
EllagitanninsChingiitannin ARubus chingii Hu
(unripe fruit/n-BuOH fraction)
4.52 ± 0.30 μM35.71 ± 4.93 μM[151]
Lambertianin A10.32 ± 0.10 μM
Sanguiin H-611.00 ± 0.21 μM
Gallotanins1,2,3,4,6-penta-O-galloyl-β-D-glucopyranoseRhus coriaria
(leaves)
6.32 ± 0.18 μM10.69 ± 0.50 μM[70]
Dihydrostilbene glycosidesSasastilboside ACamellia sasanqua Thunb
(leaves)
53.7 ± 1.6 μM-[152]
ProanthocyanidinsChinese bayberry leaves proanthocyanidinsMyrica rubra Sieb. et Zucc.
(leaves)
3.075 ± 0.073 μg/mL-[153]
PhenolicDehydrodieugenol BOcimum tenuiflorum29.6 μM13.85 μM[154]
CoumarinHalfordinMelicope latifolia197.53 µM282.39 ± 8.14 µM[71]
Alkaloids3,3′,5,5′,8-pentamethyl-3,3′-bis(4-methylpent-3-en-1-yl)-3,3′,11,11′-tetrahydro-10,10′-bipyrano[3,2-a]carbazoleMurraya koenigii (L.)30.32 ± 0.34 ppm-[155]
BerberineCardiospermum halicacabum72% at 10 μg/mL [156]
Pregnane glycosidesDrevoluoside QDregea volubilis
(leaves)
51.3 ± 2.1 µM36.3 ± 0.5 µM[157]
Gymsyloside BGymnema sylvestre
(leaves)
175.8 ± 2.3 µM72.4 ± 0.8 µM[158]
Gymsyloside C162.2 ± 2.7 µM
Gymsyloside D113.0 ± 0.7 µM
Prenylated xanthonesMangoxanthone AGarcinia mangostana
(pericarp)
22.74 ± 2.07 µM-[159]
XanthoneGarcixanthone DGarcinia mangostana
(pericarp)
93.8%96.4%[160]
Garcinone E85.6%
XanthophyllFucoxanthinPhaeodactylum tricornutum28.38 ± 0.67 mmol/L25.01 ± 1.38 mmol/L[161]
Fatty acid5,7-dihydroxy-6-oxoheptadecanoic acidTiliacora triandra26.27 ± 1.11 μM.177.65 ± 0.88 μM[162]
Phytosterolsβ-SitosterolParthenium hysterophorus
(leaves)
42.30% (at 400 µg/mL)-[163]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kashtoh, H.; Baek, K.-H. New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects. Plants 2023, 12, 2944. https://doi.org/10.3390/plants12162944

AMA Style

Kashtoh H, Baek K-H. New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects. Plants. 2023; 12(16):2944. https://doi.org/10.3390/plants12162944

Chicago/Turabian Style

Kashtoh, Hamdy, and Kwang-Hyun Baek. 2023. "New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects" Plants 12, no. 16: 2944. https://doi.org/10.3390/plants12162944

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