Next Article in Journal
Kaempferia parviflora Extracts Protect Neural Stem Cells from Amyloid Peptide-Mediated Inflammation in Co-Culture Model with Microglia
Previous Article in Journal
Mid-Luteal Progesterone Is Inversely Associated with Premenstrual Food Cravings
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Legume-Derived Bioactive Peptides in Type 2 Diabetes: Opportunities and Challenges

Sino-German Biomedical Center, Hubei Provincial Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(5), 1096; https://doi.org/10.3390/nu15051096
Submission received: 7 February 2023 / Revised: 17 February 2023 / Accepted: 20 February 2023 / Published: 22 February 2023

Abstract

:
Diabetes mellitus is a complex disorder characterized by insufficient insulin production or insulin resistance, which results in a lifelong dependence on glucose-lowering drugs for almost all patients. During the fight with diabetes, researchers are always thinking about what characteristics the ideal hypoglycemic drugs should have. From the point of view of the drugs, they should maintain effective control of blood sugar, have a very low risk of hypoglycemia, not increase or decrease body weight, improve β-cell function, and delay disease progression. Recently, the advent of oral peptide drugs, such as semaglutide, brings exciting hope to patients with chronic diabetes. Legumes, as an excellent source of protein, peptides, and phytochemicals, have played significant roles in human health throughout human history. Some legume-derived peptides with encouraging anti-diabetic potential have been gradually reported over the last two decades. Their hypoglycemic mechanisms have also been clarified at some classic diabetes treatment targets, such as the insulin receptor signaling pathway or other related pathways involved in the progress of diabetes, and key enzymes including α-amylase, α-glucosidase, and dipeptidyl peptidase-IV (DPP-4). This review summarizes the anti-diabetic activities and mechanisms of peptides from legumes and discusses the prospects of these peptide-based drugs in type 2 diabetes (T2D) management.

1. Introduction

Diabetes is a metabolic disease characterized by a persistent over-normal level of blood glucose that causes impressive morbidity and mortality worldwide [1]. Persistent hyperglycemia imposes damage on other organs, such as the eye, heart, kidney, and skin, as well as the nervous system, and is strongly correlated with a myriad of diabetes-related complications [2]. Diabetes typically develops when the pancreas does not create enough insulin, or the organs involved in glucose metabolic regulation do not effectively respond to the insulin [3]. Type 1 diabetes (T1D, 5–7%), type 2 diabetes (T2D, 90%), and gestational diabetes (GD, 2–3%) are the three most common forms of diabetes [4]. T1D is a destructive autoimmune disease of pancreatic β-cells, leading to a deficiency of insulin. T2D is the most prevalent kind of diabetes in the general population, and is frequently caused by inadequate insulin production or insulin resistance [5,6]. GD is another type of diabetes that is caused by abnormal glucose tolerance and diabetes during pregnancy [7,8]. The global prevalence of diabetes shows that 537 million adults were troubled by diabetes in 2021, and the diabetic population is estimated to rise to 643 million (11.3%) in 2030 and to 783 million (12.2%) in 2045 [9]. In recent years, the incidence of T2D cases has accelerated rapidly, and it is predicted that the T2D cases may reach 700 million in 2045 [9].
Given the grim reality of T2D, several strategies including appropriate exercise, reasonable diet, and pharmacotherapy have been applied to control postprandial hyperglycemia to prevent the development of the disease. However, pharmacotherapy is the most effective method, and is favored by T2D patients. Up to now, existing drugs such as insulin and its analogues, metformin, sulfonylurea, glucagon-like peptide-1 (GLP-1) receptor agonist, dipeptidyl peptidase IV (DPP-4) inhibitor, sodium-glucose cotransporter-2 (SGLT-2) inhibitor, α-glucosidase inhibitor, etc., have been approved for the treatment of T2D. However, because no drug has yet been identified to cure T2D, nearly all T2D patients have a lifelong dependence on glucose-lowering medications. It is worth noting that recent studies have highlighted that pretreatment via the intake of special food can significantly reduce the prevalence of T2D for high-risk groups, resulting in the minimalization of the economic impact of T2D treatment [8,10].
Biopeptides are easily absorbed food-derived protein segments that play crucial roles in controlling or pretreating metabolism-related chronic diseases, such as the mild T2D subtype [11]. Recently, legume-derived peptides, such as native peptides and hydrolytic peptides from legume proteins, have been reported to prevent or treat T2D in animal models and in clinical studies. Mechanically, these peptides regulate glucose metabolism via several ways, e.g., increasing insulin secretion, improving the sensitivity of the body’s response to insulin, and inhibiting the activities of key enzymes (α-amylase, α-glucosidase, DPP-4) [12]. Aglycin family members, which are highly conserved peptides containing 37 amino acids extracted from soybeans, can resist gastric and intestinal protease degradation and exhibit insulin-like activities [13]. Legume protein hydrolysates (also called oligopeptides), always containing 2–20 amino acids produced by enzymatic hydrolysis, microbial fermentation, or food processing, have potential anti-diabetic activity because they interact with the catalytic site of the key enzymes [12,14,15]. Researchers are increasingly concerned with the development of legume-derived hypoglycemic peptide drugs and oral-specific medical foods for the control and treatment of T2D. This review summarizes the anti-diabetic activity and hypoglycemic mechanism of legume-derived peptides, which provides an option for the development of oral hypoglycemic drugs.

2. T2D and Its Therapy

2.1. The Aberrant Glucose Metabolism in T2D

The normal regulation of glucose metabolism is determined by a feedback loop consisting of pancreatic β-cells and insulin-responding tissues (such as the liver, skeletal muscle, and adipocytes) [5]. Pancreatic β-cells, located in the islets of Langerhans, lower postprandial blood glucose through glucose-dependent insulin secretion [16]. Insulin stimulates the liver, skeletal muscles, and adipocytes to transform glucose, amino acids, and fatty acids for energy storage. When insulin resistance occurs, the insulin-responding tissues and organs cannot sensitively utilize glucose. Hence, more insulin is needed to maintain the standard glucose tolerance, which may cause the overwork and dysfunction of islet β-cells [5]. β-cells’ dysfunction results in a relative lack of insulin secretion, which causes the development of hyperglycemia or T2D [6,17].
T2D is characterized by a relative lack of insulin secretion or insulin resistance occurring in the insulin-responding organs. The current multiple drugs of T2D work mainly by reversing several physiological indexes to lower blood glucose, including an increase of insulin secretion (pancreas islet β-cells), a decrease of glucagon secretion (pancreas islet α-cells), an improvement of neurotransmitter dysfunction (brain), a decrease of hepatic glucose production (liver), an increase of peripheral glucose absorption and deceleration of lipolysis (muscle and adipose tissue), a decrease of renal reabsorption of glucose (kidney), an increase of incretin effect and decrease of glucose absorption (intestinal), an improvement of balance of gut microbiota (colon) [18]. An early start and a combination of multiple therapy drugs can bring benefits to T2D patients.

2.2. Clinical Therapeutic Drugs and Their Targets in T2D

Some anti-diabetic drugs targeting specific tissues and organs have been approved. The definite targets of these drugs are mainly α-amylase, α-glucosidase, dipeptidyl peptidase-4 (DPP-4), glucagon-like peptide-1 (GLP-1), gastric inhibitory peptide (GIP), gastrointestinal target hormone peptide (PYY), glucose transporters (GLUTs), and sodium-dependent glucose cotransporters (SGLTs) (Table 1) [5,19,20,21]. Insulin and its analogs are the most common injectable diabetes drugs for patients with severe pancreatic dysfunction. Oral anti-diabetic drugs mainly include metformin, sulfonylureas, glinides, thiazolidinediones (TZDs), DPP-4 inhibitors, SGLT-2 inhibitors, α-amylase inhibitors, and α-glucosidase inhibitors. The current medications can lessen the harm to the body brought on by hyperglycemia by regulating blood sugar levels and increasing insulin sensitivity [22].
Insulin, the most important regulator of metabolism, reduces blood sugar by preventing hepatic glucose synthesis and promoting intracellular glucose uptake by fat and muscle cells. Insulin and its analogues have a short action period and a risk of minimally invasive infection and other side effects, such as hypoglycemia, resistance, allergy, edema, and lipodystrophy [23]. The market for peptide-based medicines has been firmly established by the creation of human insulin utilizing recombinant technology, as well as other synthetic hormones [4]. Oral insulin is superior to other forms of insulin delivery because it follows the normal physiological pathway, that is, through the portal vein circulation of the liver, first reaching the liver and later the peripheral tissues [24].
Metformin, an oral medication, is used to reduce blood sugar levels in people with non-insulin-dependent diabetic mellitus. It promotes the uptake and utilization of glucose in adipocytes and muscles and improves insulin sensitivity by reducing glucose synthesis in the liver [12]. Metformin has good security, but 5–15% of the administrated patients are still troubled by side effects, such as diarrhea, nausea, indigestion, and other gastrointestinal symptoms [20]. TZDs also ameliorate insulin resistance in adipocytes, liver, and muscle [25]. Rosiglitazone and pioglitazone, peroxisome proliferator-activated receptor c (PPARc) agonists of TZDs, act as insulin sensitizers that bring benefits by maintaining long-term glycemic balance [25]. Presently, TZDs are rarely considered for the treatment of T2D due to side effects, such as cardiac issues, edema, and weight gain.
Sulfonylureas are currently used as second-line drugs for T2D or in combination with other medications. They promote insulin secretion by blocking K+ channels in pancreatic tissue, limiting gluconeogenesis, reducing the insulin clearance rate in the liver, and reducing fatty acid decomposition [26,27]. First-generation and second-generation sulfonylureas are the two categories. The tolbutamide and chlorpropamide sulfonylureas are first-generation sulfonylureas. Glyburide (also known as glibenclamide), glipizide, glimepiride, and gliclazide are second-generation sulfonylureas. These days, second-generation sulfonate drugs are used in clinical settings more frequently. All sulfonyl medications have a common side effect called hypoglycemia, along with less serious side effects like headache, nausea, dizziness, allergic reactions, and weight gain [6]. Meglitinides, non-sulfonylureas insulin secretagogues, also target pancreatic β-cell tissue, stimulating the pancreatic β-cells to secrete insulin, without hypoglycemic risk [26].
SGLT-2 inhibitors are oral hypoglycemic medications that lower blood sugar by blocking renal glucose reabsorption. They prevent about 90% of the filtered glucose reabsorption by directly inactivating the SGLT-2, a glucose transporter located in the proximal tubules’ first section [20,28]. SGLT-2 inhibitors include dapagliflozin, canagliflozin, empagliflozin, ertugliflozin, and sotagliflozin. They also have side effects, such as urinary and genital infection risk (6.4%) and ketoacidosis risk [6]. Glucose reabsorption is blocked in the kidney when SGLT-2 is inhibited, resulting in insulin-independent glucose lowering as well as a reduction in weight and blood pressure [20].
GLP-1 receptor agonists, GLP-1 analogues, and incretin mimics are common hypoglycemic drugs. Injectable GLP-1 receptor agonists include exenatide, liraglutide, lixisenatide, albiglutide, and dulaglutide. They control the blood glucose balance via the enhancement of insulin release and suppression of excessive glucagon secretion by activating the GLP-1 receptor [7]. Exedin-4 is one of the most promising GLP-1 receptor agonists currently utilized to treat diabetes, and is injected twice daily [29]. Liraglutide is a once-daily GLP-1 analogue, whose homology of amino acids sequence with human GLP-1 is 97% [30]. The modification of a fatty acid at position 26, and the replacement of lysine with arginine at position 34, result in an extension of the plasma half-life of liraglutide (13 h), an increase of self-association, and a resistance to digestion of DPP-4 [31,32]. In 2019, semaglutide (Rybelsus®), the first oral hypoglycemic peptide drug, was approved by the U.S. Food and Drug Administration (FDA) to improve glycemic control in adults with T2D [33]. The combination of semaglutide and a pharmaceutical sodium 8-[(2-hydroxybenzoyl)amino]octanoate (SNAC), an absorption enhancer, leads to a better solubility of semaglutide and an increased resistance to degradation at a low pH value [31]. Despite being combined with SNAC, the oral bioavailability of Rybelsus® is only 0.4–1% [30].
DPP-4 inhibitors are oral medications that extend the half-lives of endogenous incretins by preventing the DPP-4 enzyme from digesting incretins [28,34]. These medications include sitagliptin, vildagliptin, saxagliptin, linagliptin, and alogliptin. The hypoglycemic effect of DPP-4 inhibitors is characterized by glucose concentration dependence. Therefore, they generally may not cause hypoglycemia.
Mounjaro (tirzepatide) is a “first-in-class” injectable drug that activates both GIP and GLP-1 receptors, controlling hyperglycemia through a dual mechanism of action [35,36]. Imeglimin, another “first-in-class” oral tablet, can improve insulin secretion disorders with a novel mechanism of action targeting mitochondrial bioenergetics [37]. It rebalances respiratory chain activity, resulting in less reactive oxygen species formation and the prevention of mitochondrial permeability transition pore opening [37]. Chiglitazar, a PPAR complete agonist, is a new oral drug approved by China’s national medical products administration for T2D treatment in 2021; it can activate three subunits of PPAR (α, γ, and δ) [38,39].
α-glucosidase inhibitors (acarbose, miglitol, voglibose) and α-amylase inhibitors are the first-line drugs for T2D. They lower blood glucose levels by slowing down the digestion rate and inhibiting intestinal glucose absorption [40]. Their main side effect is gastrointestinal reaction.
Over the past few decades, there has been a significant increase in therapy alternatives, but none of them has prevented the decline of β-cell function. The current drugs available to treat T2D listed in Table 1 have side effects, and most of them are administered by injection. As a result, many scientists have made efforts to create alternative delivery methods, such as transdermal, oral, nasal, and ocular delivery. Due to the higher safety and compliance, the oral route remains the most appealing option for T2D patients. Currently, various oral peptide drugs have reached clinical trials, including oral insulin, calcitonin, parathyroid hormone, and vasopressin. The approval of an oral formulation of the GLP-1 receptor agonist semaglutide for the treatment of T2D represents a significant landmark in the development of oral peptide drugs [33].

3. Legume-Derived Peptides and Their Anti-Diabetic Activity

The legume is the fruit or seed of plants of the legume family, which are used for food, including lentils (Lens culinaris), common beans (Phaseolus vulgaris), broad beans (Vicia faba), dry peas (Pisum sativum), chickpeas (Cicer arietinum), cowpeas (Vigna sinensis), and mung beans (Vigna radiata) [41]. Legumes are an excellent source of good quality protein (20–45% protein), providing high amounts of the essential amino acids such as lysine and leucine, and lower amounts of methionine and tryptophan, complex carbohydrates, dietary fiber, unsaturated fats, vitamins, and essential minerals for the human diet [42,43].
The intake of legumes brings many benefits and positive effects for physical health, especially on metabolic syndromes, such as cardiovascular protection and the regulation of glucose and lipid metabolism [11,44]. Legume proteins can be converted into a variety of peptides that control vital biological activities, especially those involved in the endocrinology of living organisms [45,46]. Several observations of clinical statistics indicate that intake of legumes (lentils, chickpeas, and soybeans) or their protein significantly lowers the risk of T2D among Caucasians and Xanthoderms in different countries [47,48,49,50]. The relationship between the risk of T2D and the intake of legumes is listed in Table 2.
Given that legumes and their proteins benefit T2D patients, various legume-derived peptides are identified as anti-hyperglycemia agents due to their metabolic regulating bioactivity in vivo or in vitro. Plant-derived peptides have been shown to have hypoglycemic effects as well, which overcome security hurdles and yield an efficient treatment for T2D [4,51]. These agents can be divided into native proteins, peptides, and protein hydrolysates (hydrolytic peptides) [4,52]. The native proteins and peptides can be directly extracted and purified from legume storage or germinating seeds. Hydrolytic peptides, also called oligopeptides, are a mixture derived from the legume protein by digestion of various enzymes, fermentation of microorganisms, or mechanical cutting. Several studies have demonstrated the potential anti-diabetic activity of native preparations and hydrolysates in vitro, in vivo, or in clinical trials [53,54,55].
Animal models are the most common approaches to investigate the anti-diabetic activity of drugs in vivo, including streptozotocin (STZ) and high-fat diet (HFD)-induced diabetic mice [14,56,57], KKAy diabetic mice [58], T1D Sprague Dawley (SD) rats [14,59], STZ-induced Wistar rats [60,61], HFD-C57BL/6J mice [15], and alloxan-induced diabetic mice [62]. The potential anti-diabetic benefits of legume hydrolysates and fractions or synthesized pure peptides have been investigated by several cell line models. Glucose consumption activities were evaluated by human HepG2 [15,57,59,63,64], Caco-2 [64], 3T3-L1 [64,65], and INS-1E cells [14,66]. Furthermore, Rat L6 skeletal muscle cells [67], Min6 cell NCI-H716 [65], and human embryo kidney 293 cell models [65] were used to evaluate the activation of the insulin signaling pathway. The legume-derived peptides related to anti-diabetic activities, experimental models, and anti-diabetic mechanisms are summarized in Table 3.

3.1. Native Legume Peptides

The primary elements of legume seeds are proteins and peptides generated by the connection of amino acids via peptide bonds. Native legume peptides (aglycin, vglycin and soymorphin-5) have been reported as anti-T2D agents due to their bioactivities in decreasing blood sugar levels, improving insulin sensitivity and glucose tolerance, and promoting the proliferation of β-cells (Table 3).
Aglycin, a biopeptide containing 37 amino acid residues, was identified as matching to the residues 27–63 of plant albumin 1 B precursor (PA1B) from pea seeds [56,77]. Aglycin has a high affinity with a 43 kDa basic 7S globulin protein [78]. This protein binding mode is similar to the interaction of insulin and insulin receptor; therefore, aglycin is also called leginsulin [79]. Structurally, it is made up of six cysteines embedded in three disulfide bonds (C3-C20, C7-C22, and C15-C32), which are highly conserved and can resist the degradation of pepsin, trypsin, and Glu-C protease [56,57]. Aglycin was initially thought to be a leginsulin that participates in plant signal transduction to control growth and differentiation [80]. Subsequently, Chen’s group found that the oral administration of aglycin in diabetic mice effectively controlled hyperglycemia by improving the insulin-signaling pathway and enhancing glucose uptake in peripheral tissues [77]. Furthermore, a long-term administration of aglycin benefited the impaired pancreatic restoration and insulin secretion [56].
Vglycin, another leginsulin, is highly similar to aglycin in amino acid sequence and structural domain [57]. It may function with a similar mechanism to aglycin, which could be useful in T2D treatment for restoring impaired insulin signaling, glucose tolerance, and pancreatic function [14]. Other leginsulin variants identified in different cultivars, as well as their bacteria-expressed recombinants, also exert insulin-like activities in myotube-like differentiated L6 and C2C12 cells [13]. Thus, leginsulins, such as aglycin and vglycin, could be considered as potential orally administrated drugs to be developed.
Soymorphin-5 (YPFVV), a µ-opioid agonist with five amino acids, is derived from soybean β-conglycinin β-subunit, and has anxiolytic-like activity [58]. It exhibited good glucose lowing activity in diabetic KKAy mice [58]. Soymorphin-5 lowered blood glucose levels in the glucose tolerance test (GTT) and insulin tolerance test (ITT) after acute intraperitoneal administration, implying that soymorphin-5 might increase insulin sensitivity in KKAy mice [58]. Soymorphin-5 benefits diabetic improvement by increasing plasma insulin levels and decreasing glucose and triglyceride (TG) levels. However, des-Tyr-soymorphin-5 (PFVV) loses the µ-opioid and anxiolytic-like activities, as well as their metabolic regulating activity [58].

3.2. Legume Proteins

Legume seed proteins are the main dietary source of plant protein, in which anti-nutritional substances include lectins, hydrolase inhibitors, ribosome-inactivating proteins (RIPs), and allergens [81]. Recently, soy and lupin proteins have been reported to have hypoglycemic effects.
Most of the protein in lupin seeds (around 44% w/w of the total conglutins) belongs to the conglutin family, whose molecular mass is between 143 and 260 kDa [82]. β1-, β3-, and β6-conglutin proteins can bind to insulin in vitro, suggesting that β-conglutin proteins have potential benefits for the prevention and treatment of diabetes [83]. The second most prevalent globulin, α-conglutin, shares 33% w/w of all conglutins. The third conglutin is δ-conglutin, which accounts for 12% w/w of all conglutins and 4–5% w/w of all globulins [82]. γ-conglutin (~50 kDa) is a glycoprotein composed of two subunits (17 kDa and 29 kDa). γ-conglutin has excellent hypoglycemic effects in vitro and in vivo (Table 3). In mouse C2C12 cells, γ-conglutin activated the kinases of the synthetic protein pathway and increased glucose consumption by up-regulating muscle-specific gene MHC transcription and GLUT4 gene translocation [73]. In the STZ rat model, Ins1 gene expression and pancreatic insulin content were elevated by γ-conglutin [72]. Bertoglio’s group reported that lupin γ-conglutin had a glucose-lowering effect in a clinical trial [54]. When different dosages of γ-conglutin (630, 315, and 157.5 mg) were eaten 30 min before the carbohydrate supply for 7 weeks, a relevant hypoglycemic effect could be observed in human subjects [54]. Lupin seed protein could be transported by Caco-2 cell monolayers and everted intestinal sacs, suggesting it has the potential to be developed as an oral agent to manage T2D [84].
Soybean storage proteins, such as β-conglycinin and glycinin, also promote anti-diabetic activity (Table 3). Nordentoft reported that soy protein improved insulin sensitivity and up-regulated the expression of key insulin-regulatory genes [69]. A clinical study compared the effects of the intake of 6% (w/v) whey protein isolate (WPI), whey protein hydrolysate (WPH), soy protein isolate (SPI), and 2.66% WPI, or a control (no protein added) on insulin and glucose responses in 25 healthy men. The results suggested that SPI with 6% protein enhanced insulin response and decreased the plasma glucose level compared with a control beverage [55]. Another study demonstrated that the SPI increased the insulin response compared to WPH [85].

3.3. Hydrolytic Peptides

Bioactive hydrolytic peptides are organic substances formed from amino acids and released mainly through enzymatic proteolysis or food processing (cooking, fermentation, ripening) [86,87]. Currently, the commonly used peptide and protein extraction methods mainly include aqueous, organic solvent, enzymatic, ultrasonic, two-phase aqueous, and reverse micelle extraction [86]. The two most common methods for making bioactive peptides are enzyme hydrolysis and fermentation [88]. Pepsin, trypsin, chymotrypsin, alcalase, and flavourzyme are frequently utilized for hydrolysis. Compared with microbial fermentation, these bioactive oligopeptides, occurring in different sizes and produced by enzymes, have higher scalability [4]. The peptides, after enzymatic digestion, can be recovered by desalination, membrane ultrafiltration, column chromatography, and freeze-drying [87].
Crucially, some oligopeptides with short sequences of 2–20 amino acids can be absorbed by the intestine into the blood circulation and exert systemic or local physiological anti-diabetic activities, such as decreasing blood glucose levels, improving glucose and insulin sensitivity in target tissues, and inhibiting key enzymes like α-amylase, α-glucosidase, and DPP-4 related to T2D [4,89,90].
Legume-derived hydrolysates and fractions have also been investigated in cell and animal models (Table 3). Administered with semipurified γ-conglutin, hydrolysates (5 mg/mL) from Andean lupin legumes induced a 6.5-fold glucose uptake and decreased gluconeogenesis in male SD rats by 50% [64]. In Caco-2 cells, treatment with 10 mg/mL black bean hydrolyzed protein isolate (HPI) decreased glucose absorption by 21.5%, and intake of 100–200 mg of HPI/kg BW/day lowered postprandial glucose levels by 22.7–47.7% in a hyperglycemic rat model [60]. The intake of common bean peptide fractions reduced glucose levels in male Wistar rats at the same rate as glibenclamide, keeping a stable basal level without a surge in postprandial hyperglycemia [75]. Jiang found that soybean protein hydrolysate (<5 kDa) made by ultrafiltration significantly reduced the level of fasting blood glucose (FBG) in mice [62]. Wei reported that pea oligopeptides significantly reduced blood glucose levels, lipid profiles, and liver fat deposition in diabetic mice [74].
Some pure sequence-known peptides have also shown hypoglycemic effects in animal and cell models (Table 3). LPYP, IAVPGEVA, and IAVPTGVA from soy glycinin improved glucose metabolism by increasing glucose uptake via up-regulating the expression of GLUT1 and GLUT4 in cultured hepatic cells [63]. Lammi’s group developed a fast, sensitive, and cost-effective ex vivo DPP-4 assay for human serum by collecting venous blood from a healthy female volunteer and analyzing how peptides inactivated the enzyme. They discovered that hydrolytic peptides like LTFPGSAED from lupin β-conglutin and IAVPTGVA from soybean glycinin inhibited the activity of DPP-4 [91].
Furthermore, legume pure peptides and hydrolysates have been found to inhibit key enzymes (α-amylase, α-glucosidase, and DPP-4) in vitro or in silico. Herein, we summarize 40 peptides with clear amino acid sequences and their indicated targets (Table 4). A total of 19 kinds of peptides from the pinto bean, black bean, hard-to-cook bean, chickpea, and fermented bean seeds have been found to inhibit the activities of α-amylases (in biochemical assay or in silico); their sequences, sources, mass weights, and IC50 values are listed in Table 4. In the chemical assay, pinto bean peptides exhibited excellent IC50 values in inhibiting α-amylase. LSSLEMGSLGALFVCM obtained the lowest IC50 value of 0.31 mM compared with others, such as PLPLHMLP (5.92 mM), PPMHLP (6.08 mM), PPHMGGP (6.14 mM), PLPWGAGF (6.64 mM), and PPHMLP (23.33 mM) [92,93]. Some α-amylase inhibitory peptides were identified in fermented bean seeds, such as INEGSLLLPH FVVAEQAGNEEGFE, SGGGGGGVAGAATASR, GSGGGGGGGFGGPRR, GGYQGGGYGGNSGGGYGNRG, GGSGGGGGSSSGRRP, and GDTVTVEFDTFLSR. Their IC50 values ranged from 0.04 μg/mL to 0.65 μg/mL [94]. In in silico study, some peptides seemed to provide potent inhibition against α-amylase, α-glucosidase, and DPP-4 by computational modelling. Compared with acarbose (−9.71 kcal/mol), a known α-amylase inhibitor, the free energy (interaction with the catalytic site of α-amylase) of peptides AKSPLF and WEVM were −10.2 kcal/mol and −10.1 kcal/mol, respectively [95]. QQEG from hard-to-cook beans inhibited α-amylase, and the interaction score was −7.29 REU [66]. Twelve peptides provided the potential inhibition of α-glucosidase, including aglycin, LLPLPVLK, SWLRL, WLRL, GSR, EAK, TTGGKGGK, KKSSG, GGGLHK, CPGNK, KTYGL, and YVDGSGTPLT (Table 4). The α-glucosidase inhibitory activity of aglycin from soybeans was 36.48 μmol/L [96]. GSR and EAK, identified from soybeans, had IC50 values of 20.40 µM and 520.20 µM, respectively [62]. LLPLPVLK, SWLRL, and WLRL from soy protein showed strong α-glucosidase inhibitory activity, with IC50 values of 237.43 ± 0.52 µM, 182.05 ± 0.74 µM, and 162.29 ± 0.74 µM, respectively [97]. Four synthesized peptides (KKSSG, GGGLHK, CPGNK and KTYGL) were evaluated with the α-glucosidase inhibitory ratio of 49.34 ± 6.5%, 46.10 ± 8.30%, 37.60 ± 6.8%, and 36.30 ± 8.80%, respectively [98].
Peptides from the lupin bean, soybean, fermented soybean, black bean, common bean, chickpea, and hard-to-cook bean exhibited inhibition of DPP-4. Peptides LTFPGSAED, IAVPTGVA, EGLELLLLLLAG, AKSPLF, FEELN, KKSSG, GGGLHK, CPGNK, KTYGL, KL, and LR were confirmed to inhibit the activity of DPP-4 by biochemical assay. RGPLVNPDPKPFL, PHPATSGGGL, YVDGSGTPLT, LLSL, and QQEG were identified as potential DPP-4 inhibitors by computational modelling (Table 4).
In addition, several studies showed that a hydrolysate and peptide fraction mixture had the potential to inhibit the enzymatic activities of α-amylase, α-glucosidase, and DPP-4 (Table S1). Alkaline protease was reported to be the best enzyme for producing black bean protein components with anti-diabetic potential [60,95]. Inhibition values for DPP-4, α-amylase, and α-glucosidase of alcalase fraction peptides from the black bean were 96.70%, 53.40%, and 66.10%, respectively [95]. The inhibition of α-glucosidase in different common bean cultivars’ protein hydrolysates ranged from 46.90–60.00% [101], and the IC50 of DPP-4 inhibition ranged from 0.14–0.33 mg DW/mL [98]. In another in vitro study, 5 mg/mL purified γ-conglutin hydrolysates from lupin inhibited DPP-4 completely. The fact that undigested γ-conglutin did not directly inhibit DPP-4 activity demonstrated that this protein needs to be digested to produce DPP-4 enzyme inhibitory hydrolysates [64]. Bambara bean hydrolysates produced by alcalase and thermolysin exhibited similar DPP-4 inhibitory activity with an IC50 of 1.73 mg/mL [102]. Cowpea alcalase protein hydrolysis also showed strong DPP-4 inhibition [103,104]. Soybean hydrolysates had an α-glucosidase inhibitory activity with an IC50 of 0.05 mg/mL [62]. Six-days-germinated soybean protein hydrolysates inhibited DPP-4 (IC50 = 1.49 ± 0.14 mg/mL), α-amylase (IC50 = 1.70 ± 0.18 mg/mL), and sucrase activities of α-glucosidases (IC50 = 2.90 ± 0.07 mg/mL). Peptide fractions of 5–10 kDa and >10 kDa were more effective at inhibiting DPP-4 (IC50 = 0.91± 0.17 mg/mL and 1.18 ± 0.15 mg/mL, respectively) [61].
Collectively, legume peptides and hydrolysates show remarkable hypoglycemic effects through various approaches. Interest has been directed toward the acceptable oral administration of bioactive peptides, one of the main attractions for T2D patients. Based on their inherent amino acid composition and sequence, bioactive peptides exhibit different activities in regulating glucose and lipid metabolism, which exert beneficial effects on the control of diabetic progress.

4. Potential Hypoglycemic Mechanism of Bioactive Legume Peptides

The hypoglycemic mechanism of legume diabetic peptides mainly includes reducing glucose absorption, promoting pancreatic β-cells proliferation, enhancing insulin secretion and sensitivity through signaling pathways associated with diabetes, and inhibiting carbohydrate-digesting enzymes (α-amylase and α-glucosidase) and DPP-4 in target organs (Figure 1).

4.1. Targeting the Pancreas

One of the main factors contributing to the development of T2D is assumed to be a decrease in glucose-stimulated insulin secretion from pancreatic β-cells [105]. An appealing idea for treating diabetes and its complications is to restore the function of poor β-cells. The IRS (insulin receptor substrate)/AKT (protein kinase B) pathway is the primary mechanism of the hypoglycemic effect of the body’s response to insulin stimulation [106]. Vglycin peptide exhibited beneficial effects on hyperglycemia by facilitating glucose metabolism and enhancing insulin sensitivity, but it could not increase insulin secretion [57]. Another study supports an apparent impact of vglycin on regulating β-cell preservation; a self-renew program was initiated in which vglycin directly promoted the proliferation of β-cells via activating the IR/Akt/Erk pathway [14]. These results reveal the protective effects of vglycin on improving β-cell function in long-term glucolipotoxicity.
Some hydrolysates and fractions also exhibited a protective effect on the dysfunctional pancreas. Hydrolysates (<3 kDa) from hard-to-cook beans and common beans were reported to increase glucose-stimulated insulin secretion up to 57% and 45% from the basal state in INS-1E pancreatic β-cells, respectively [66,76]. In another study, fermented meju water extracts (W-60), containing 15 kDa peptides, increased glucose-stimulated insulin secretion capacity and β-cell viability in Min6 insulinoma cells [65]. Previous studies have identified that amino acid metabolism, especially glutamine and alanine, is essential for the functional maintenance of pancreatic cells and insulin secretion [107]. Therefore, it can be inferred that the amino acids or oligopeptides in the hydrolysates of legumes may be beneficial for the exertion of hypoglycemic function of the pancreas directly or indirectly.

4.2. Targeting the Liver

Soymorphin-5 (YPFVV), derived from the soy conglycinin subunit, plays a role in glucose and lipid metabolism [58]. Although no differences were observed in the weights of mesenteric and epididymal fat and brown adipose tissues, plasma TG levels and liver weights in KKAy mice were significantly reduced by soymorphin-5 treatment, suggesting that soymorphin-5 enhanced the liver lipid metabolism. In terms of mechanism, soymorphin-5 improved fatty acid β-oxidation and energy expenditure by increasing the hepatic expression of PPAR and its target genes, including Adipor2, AOX1, CPT1, and UCP2 [58]. In another study, vglycin was probably responsible for increased fatty acid β-oxidation via the AMPK pathway and inhibited fatty acid synthesis (FAS) by down-regulating expression of FAS in HFD-fed C57BL/6J mice [15]. Soy peptides IAVPGEVA, IAVPTGVA, and LPYP enhanced glucose uptake through GLUT1 and GLUT4 activation, and modulated glucose metabolism by activating Akt and AMPK pathways in HepG2 cells [63]. In addition, γ-conglutin hydrolysates decreased gluconeogenesis by 50% in a dual-layered enterocyte/hepatocyte system and down-regulated the expression of phosphoenolpyruvate carboxykinase (PEPCK) in HepG2 cells [64].

4.3. Targeting Muscle and Adipose Tissue

Increasing glucose absorption and transformation in muscle and adipose tissue is an effective method to control hyperglycemia. As shown in a STZ/HFD-induced diabetic mice model, oral administration of aglycin could potentially attenuate or prevent hyperglycemia via enhancing the IR/IRS1 signaling pathway in skeletal muscle [56]. Additionally, in C2C12 cells, aglycin increased glucose uptake by recruiting glucose transporter GLUT4 to the surface of cells [56]. Cowpea peptides induced the Akt phosphorylation in skeletal cells and mimicked the actions of insulin by activating the same signaling pathway [67]. Prior findings strongly implied that the translocation of GLUT4 was regulated by activation of the Akt signaling pathway in the skeletal muscle cells, which facilitated glucose uptake [108,109].
Studies of fermented soy products have suggested that the small bioactive peptides produced during fermentation may contribute to preventing or slowing the progression of T2D [65,110]. Water extracts (<3 kDa) from chungkookjang increased PPAR-r activity by about 59%, indicating that there exist certain small peptides that work as mild PPAR-r agonists in glucose and lipid metabolism [65]. γ-conglutin hydrolysates also significantly increased the insulin-induced glucose uptake in mature 3T3-L1 adipocytes through an enhanced expression of GLUT4, similar to metformin treatment [64]. Similarly, in C2C12 cells, γ-conglutin increased flottilin-2, caveolin-3 concentrations, and CBL phosphorylation, resulting in GLUT4 translocated on the cell membrane, which suggested that γ-conglutin plays an essential role in muscle energy metabolism through insulin-mimetic action [73].

4.4. Targeting the Intestine and Colon

In STZ-induced Wistar rats and the Caco-2 cell models, black bean peptides AKSPLF, ATNPL, FFEELN, and LSVSVL may have reduced glucose absorption via blocking glucose transporters GLUT2 and SGLT1 (Figure 1) [60]. In Table 4, we summarized 19 α-amylase inhibitors, 12 α-glucosidase inhibitors, and 16 DPP-4 inhibitors from legume peptides, all of which worked as potential anti-diabetic agents targeting the intestine and colon (Figure 1). The mechanism of lowering postprandial hyperglycemia by inhibition of α-amylase and α-glucosidase is to inhibit the rate of glucose release and absorption in the intestine [12]. The hypoglycemic mechanism of inhibiting the DPP-4 enzyme is to retard the decomposition of GLP-1 and increase the endogenous physiological level of GLP-1 in the human body [34]. Recently, the structure of oligopeptides and their binding modes to these enzymes have been systematically investigated, which may provide a clear anti-diabetic mechanism for these peptides [40].
α-amylase contains three distinct structural domains (A, B, and C). The active site region in domain A is associated with an extended substrate binding cleft, allowing it to recognize polymeric starch molecules [111]. Asp197, Glu233, and Asp300 are three essential catalytic carboxylic acid residues located within this active site cleft between the carboxyl ends of domains A and B. Domain B is responsible for substrate specificity and enzyme stability, and domain C helps to maintain the stability of the catalytic site. Therefore, blocking the catalytic sites was an effective way to inhibit the activity of α-amylase [40]. Peptides amply containing aromatic residues such as Leu, Pro, Gly, and Phe were effective inhibitors of α-amylase because the substrate-binding pockets of the α-amylase enzyme have many aromatic residues [111]. Hence, apart from interactions involving hydrogen bonds and electrostatic and van der Waals interactions, aromatic–aromatic interactions have also played a crucial role in α-amylase inhibitory activity [4].
The bioactive peptides LSSLEMGSLGALFVCM, PLPLHMLP, PPMHLP, PPHMLP, PPHMGGP, and PLPWGAGF, extracted from pinto beans, interacted or bound to the indicated domain of α-amylase, ultimately restricting protein conformation and affecting the degree of enclosure of the substrate. Mechanically, when the (α-amylase)–starch complex was formed, the uncompetitive peptide inhibitors bound to the complex, resulting in the starch no longer being able to hold on at its original position due to the altered conformation of α-amylase. Consequently, the starch was detached from the (α-amylase)–starch complex [92]. The competitive inhibitors GSR and EAK formed hydrogen bonds with the catalytic residues [62].
Structural analysis has clearly shown the intimate interaction between bean-derived peptides and α-amylase catalytic residues. The interaction prevented residue Asp300 from adopting its functional position and destroyed the water channel leading from the “flexible loop” to the heart of the active-site depression [112]. Another inhibitory mechanism was identified in the interaction of porcine pancreatic α-amylase and the bean P. vulgaris-derived peptide, in which residue His305 participated in substrate binding and structural changes [113].
Similar to peptides that inhibit α-amylase, certain peptides have the potential to inhibit α-glucosidase, reducing glucose absorption and delaying carbohydrate digestion, thereby reducing postprandial blood glucose levels. Pure peptides such as KTYGL were able to inhibit the enzyme mainly through polar interactions (Asn32, Asp34, Asp38, and Asp89), hydrophobic interactions (Trp36 and Trp81), and hydrogen bonds (Ser31) with the residues from the binding pocket of the α-glucosidase [98]. AKSPLF showed only hydrogen bonding and polar interactions with α-glucosidase, and Ala and Lys were the only two amino acids interacting with ASP34, THR83, ASP89 and ASN32 [95]. Besides, α-glucosidase inhibitors also played crucial roles in the secretion of GLP-1 in T2D [114].
DPP-4 is a serine exopeptidase that cleaves X-proline or X-alanine dipeptides from the N-terminus of polypeptides, which degrades and inactivates GLP-1 and GIP. Several legume-derived peptides have been identified as promising DPP-4 inhibitors, potentially contributing to glycemic control. DPP-4 has hydrophobic pockets, which are crucial targets for inhibition of the enzyme [115]. Generally, the DPP-4 inhibitory peptides often contain high concentrations of hydrophobic amino acids such as alanine, glycine, isoleucine, leucine, phenylalanine, proline, methionine, tryptophan, and valine [4]. The interactions between DPP-4 and common bean peptides (KTYGL and AKSPLF) were mainly H-bond, hydrophobic, polar and cation π bonds [85,92]. The interactions of DPP-4 with peptide AKSPLF occurred primarily between amino acids GLU191, ASP192, ARG253, and LEU235 [95].
Collectively, legume peptides exhibit great inhibitory potential due to their affinity for and specificity of action on key enzymes such as α-amylase, α-glucosidase, and DPP-4, which are promising candidates for the development of anti-diabetic drugs. However, the structure–activity relationships between peptides and these enzymes remain to be uncovered.

5. Challenges of Legume Peptides’ Development in T2D Treatment

Despite their tremendous potential value in anti-T2D therapies, legume-derived peptides have intrinsic disadvantages, including poor chemical and physical stability, poor bioavailability, short effective half-life, and inefficiency of oral delivery, which must be considered for their application as nutraceuticals or medicines [12,116]. Effectively delivering therapeutic peptides via the oral route is still a big challenge, even though some drawbacks can be solved by rational design with the increase of biological activity, selectivity, or bioavailability. Numerous studies have concentrated on creating novel technologies through physical and chemical methods, including enteric coating, enzyme inhibitors, permeability enhancers, nanoparticles, and intestinal microdevices, to improve the oral bioavailability and stability of the peptides, which overcome the absorptive barriers in the gastrointestinal tract [4,22,117].
Low selectivity, resistance to protein hydrolysis, and the demands of large-scale production are also some of the main challenges [22]. Approaches such as proteomic analysis, bioinformatics tools, pretreatments, hydrolysis mechanisms and isolation, and effective cell and animal screening models all contribute to obtaining hydrolysates or peptides with hypoglycemic activity [118]. Regarding the delivery and administration of peptides in the human body, the effectivity and stability of the peptides in the gastrointestinal tract are worth considering. Since oligopeptides are predicted to be resistant to digestive enzymes, they can be utilized as an ingredient or supplement in nutraceutical foods. Those specific peptides with long amino acid sequences or hydrolysates are still susceptible to digestive enzymes.
In addition, some legume-derived anti-diabetic peptides are mainly studied by integrated computational methods or in vitro assays, due to a lack of sufficient animal and human clinical trials. These potential anti-diabetic peptides need a deep investigation geared towards the development of nutraceuticals and drugs. On the other hand, hydrolytic peptides are generated from dietary proteins during gastrointestinal digestion or are produced by processing proteins. Their bioavailability after oral administration depends on their absorption and distribution to reach target organs [119]. The peptides in the legume hydrolysates (Table S1) should be identified, and their anti-diabetic activities and precise mechanisms need further investigation. Undoubtedly, legume-derived peptides research can benefit from pharmaceutical approaches for the discovery, design, and development of nutraceutical or oral medicinal peptides for T2D.

6. Conclusions and Perspectives

Peptides are becoming one of the most attractive candidates for the development of oral anti-diabetic drugs. The FDA has approved Rybelsus® (semaglutide) oral tablets to improve control of blood sugar in adult patients with T2D, along with diet, exercise, and other anti-diabetic drugs. The efficacy and safety of the treatment of Rybelsus®, assisted with Salcaprozate sodium, an absorption enhancer, have been considered acceptable in several clinical trials [31,120,121]. It provides a viable design for oral legume-derived drug development for T2D. The biopeptides and protein hydrolysates from legumes summarized in this review are confirmed to have anti-diabetic activities, and their anti-diabetic mechanisms are illustrated in vivo and in vitro.
The biological activity of a peptide mainly depends on its chemical structure, which determines its molecular function in target organs [44,94]. Structural modifications can enhance the enzymatic tolerance of polypeptide drugs, thereby increasing the half-life of drugs. As a native peptide, aglycin consists of 37 amino acids and contains the special cystine knot structure in the molecule, which can resist the hydrolysis of proteases in the digestive tract. This self-protective structural feature makes aglycin viable as an oral agent in the management of T2D and other metabolic syndromes.
Since its discovery, insulin has been one of the major therapeutic molecules used to control diabetes. The most common route of insulin administration is the subcutaneous route. However, oral delivery is the most convenient and patient-centered route. The human physiological system acts as a formidable barrier to insulin, limiting its bioavailability. Currently, the delivery methods enhancing the bioavailability of oral insulin and GLP-1 receptor agonists include the use of ionic liquids, hydrogels, nanoparticles, microparticles, and nano-in-microparticles [122]. These strategies provide new insights into the development of the effective delivery of orally administered protein and peptide drugs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu15051096/s1, Table S1: Hydrolytic fractions from legume targeting α-amylase, α-glucosidase, and DPP-4.

Author Contributions

Conceptualization, K.H., H.H. and C.Y.; formal analysis, K.H. and C.Y.; writing—original draft preparation, H.H.; writing—review and editing, K.H., H.H. and C.Y.; comments and suggestion, H.L. and Y.W.; supervision, K.H. and C.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Open Project Funding of the Key Laboratory of Fermentation Engineering (Ministry of Education, grant No. 202209EF09) to C.Y., by the Key R&D Project of Hubei Province (Social Development), grant No. 2022BCA018 to K.H., and by the Hubei Province Natural Science Foundation, grant No. 2021CFB289 to H.L. The APC was funded by grant No. 2022BCA018.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

With many thanks to Zhengwang Chen (Huazhong University of Science and Technology), from whose instruction on the treatment of T2D, we benefited greatly.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Aktprotein kinase B
AUCarea under curve
BGblood glucose
BWbody weight
γ-conglutin
DPP-4dipeptidyl peptidase IV
FAfatty acid
FASfatty acid synthase
FBGfasting blood glucose
FPGfasting plasma glucose
GIPgastric inhibitory peptide
GLP-1glucagon-like peptide-1
GLUT4glucose transporter type 4
HFDhigh-fat diet
IC50half maximal inhibitory concentration
IRinsulin receptor
IRS1insulin receptor substrate 1
ITTinsulin tolerance test
OGTToral glucose tolerance test
PPARperoxisome proliferator-activated receptor
REUrosetta energy units
SDSprague Dawley
SGLT-2sodium-glucose cotransporter-2
STZstreptozotocin
T2Dtype 2 diabetes
TGtriglyceride
TZDsthiazolidinediones

References

  1. Lin, X.; Xu, Y.; Pan, X.; Xu, J.; Ding, Y.; Sun, X.; Song, X.; Ren, Y.; Shan, P.F. Global, regional, and national burden and trend of diabetes in 195 countries and territories: An analysis from 1990 to 2025. Sci. Rep. 2020, 10, 14790. [Google Scholar] [CrossRef]
  2. American Diabetes Association. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2021. Diabetes Care 2021, 44 (Suppl. 1), S15–S33. [Google Scholar] [CrossRef]
  3. Al-Goblan, A.S.; Al-Alfi, M.A.; Khan, M.Z. Mechanism linking diabetes mellitus and obesity. Diabetes Metab. Syndr. Obes. 2014, 7, 587–591. [Google Scholar] [CrossRef] [Green Version]
  4. Antony, P.; Vijayan, R. Bioactive Peptides as Potential Nutraceuticals for Diabetes Therapy: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 9059. [Google Scholar] [CrossRef]
  5. 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] [Green Version]
  6. Sterrett, J.J.; Bragg, S.; Weart, C.W. Type 2 Diabetes Medication Review. Am. J. Med. Sci. 2016, 351, 342–355. [Google Scholar] [CrossRef]
  7. Acquah, C.; Dzuvor, C.K.O.; Tosh, S.; Agyei, D. Anti-diabetic effects of bioactive peptides: Recent advances and clinical implications. Crit. Rev. Food Sci. Nutr. 2022, 62, 2158–2171. [Google Scholar] [CrossRef]
  8. Hewage, S.S.; Wu, S.; Neelakantan, N.; Yoong, J. Systematic review of effectiveness and cost-effectiveness of lifestyle interventions to improve clinical diabetes outcome measures in women with a history of GDM. Clin. Nutr. ESPEN 2020, 35, 20–29. [Google Scholar] [CrossRef]
  9. International Diabetes Federation. IDF Diabetes Atlas, 10th ed. Available online: https://diabetesatlas.org/en/resources/ (accessed on 5 December 2022).
  10. Mata-Cases, M.; Rodriguez-Sanchez, B.; Mauricio, D.; Real, J.; Vlacho, B.; Franch-Nadal, J.; Oliva, J. The Association Between Poor Glycemic Control and Health Care Costs in People with Diabetes: A Population-Based Study. Diabetes Care 2020, 43, 751–758. [Google Scholar] [CrossRef]
  11. Mullins, A.P.; Arjmandi, B.H. Health Benefits of Plant-Based Nutrition: Focus on Beans in Cardiometabolic Diseases. Nutrients 2021, 13, 519. [Google Scholar] [CrossRef]
  12. Moreno-Valdespino, C.A.; Luna-Vital, D.; Camacho-Ruiz, R.M.; Mojica, L. Bioactive proteins and phytochemicals from legumes: Mechanisms of action preventing obesity and type-2 diabetes. Food Res. Int. 2020, 130, 108905. [Google Scholar] [CrossRef]
  13. Hashidume, T.; Sakano, T.; Mochizuki, A.; Ito, K.; Ito, S.; Kawarasaki, Y.; Miyoshi, N. Identification of soybean peptide leginsulin variants in different cultivars and their insulin-like activities. Sci. Rep. 2018, 8, 16847. [Google Scholar] [CrossRef] [Green Version]
  14. Jiang, H.; Tong, Y.; Yan, D.; Jia, S.; Ostenson, C.G.; Chen, Z. The Soybean Peptide Vglycin Preserves the Diabetic beta-cells through Improvement of Proliferation and Inhibition of Apoptosis. Sci. Rep. 2015, 5, 15599. [Google Scholar] [CrossRef] [Green Version]
  15. Yao, C.-C.; Tong, Y.-X.; Jiang, H.; Yang, D.-R.; Zhang, X.-J.; Zhang, P.; Su, L.; Zhao, Y.-Y.; Chen, Z.-W. Native polypeptide vglycin prevents nonalcoholic fatty liver disease in mice by activating the AMPK pathway. J. Funct. Foods 2020, 73, 104110. [Google Scholar] [CrossRef]
  16. Aguayo-Mazzucato, C.; Bonner-Weir, S. Pancreatic beta Cell Regeneration as a Possible Therapy for Diabetes. Cell Metab. 2018, 27, 57–67. [Google Scholar] [CrossRef] [Green Version]
  17. Weir, G.C.; Gaglia, J.; Bonner-Weir, S. Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes. Lancet Diabetes Endo. 2020, 8, 249–256. [Google Scholar] [CrossRef]
  18. Defronzo, R.A. Banting Lecture. From the triumvirate to the ominous octet: A new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009, 58, 773–795. [Google Scholar] [CrossRef] [Green Version]
  19. Moller, D.E. New drug targets for type 2 diabetes and the metabolic syndrome. Nature 2001, 414, 821–827. [Google Scholar] [CrossRef]
  20. Bailey, C.J. The Current Drug Treatment Landscape for Diabetes and Perspectives for the Future. Clin. Pharmacol. Ther. 2015, 98, 170–184. [Google Scholar] [CrossRef]
  21. Thule, P.M. Mechanisms of current therapies for diabetes mellitus type 2. Adv. Physiol. Educ. 2012, 36, 275–283. [Google Scholar] [CrossRef] [Green Version]
  22. Zhu, Q.; Chen, Z.; Paul, P.K.; Lu, Y.; Wu, W.; Qi, J. Oral delivery of proteins and peptides: Challenges, status quo and future perspectives. Acta Pharm. Sin. B 2021, 11, 2416–2448. [Google Scholar] [CrossRef]
  23. Yari, Z.; Behrouz, V.; Zand, H.; Pourvali, K. New insight into diabetes management: From glycemic index to dietary insulin index. Curr. Diabetes Rev. 2020, 16, 293–300. [Google Scholar] [CrossRef]
  24. Rekha, M.R.; Sharma, C.P. Oral delivery of therapeutic protein/peptide for diabetes--future perspectives. Int. J. Pharm. 2013, 440, 48–62. [Google Scholar] [CrossRef]
  25. Soccio, R.E.; Chen, E.R.; Lazar, M.A. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014, 20, 573–591. [Google Scholar] [CrossRef] [Green Version]
  26. Akter, H. Review on diabetics and anti-diabetic drugs. AJPTI 2014, 2, 1–16. [Google Scholar]
  27. DeFronzo, R.A. Pharmacologic Therapy for Type 2 Diabetes Mellitus. Ann. Intern. Med. 1999, 131, 281–303. [Google Scholar] [CrossRef]
  28. Bailey, C.J.; Tahrani, A.A.; Barnett, A.H. Future glucose-lowering drugs for type 2 diabetes. Lancet Diabetes Endo. 2016, 4, 350–359. [Google Scholar] [CrossRef] [Green Version]
  29. Furman, B.L. The development of Byetta (exenatide) from the venom of the Gila monster as an anti-diabetic agent. Toxicon 2012, 59, 464–471. [Google Scholar] [CrossRef]
  30. Liao, H.J.; Tzen, J.T.C. Investigating Potential GLP-1 Receptor Agonists in Cyclopeptides from Pseudostellaria heterophylla, Linum usitatissimum, and Drymaria diandra, and Peptides Derived from Heterophyllin B for the Treatment of Type 2 Diabetes: An In Silico Study. Metabolites 2022, 12, 549. [Google Scholar] [CrossRef]
  31. Knudsen, L.B.; Lau, J. The Discovery and Development of Liraglutide and Semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef] [Green Version]
  32. Scott, L.J. Liraglutide: A review of its use in adult patients with type 2 diabetes mellitus. Drugs 2014, 74, 2161–2174. [Google Scholar] [CrossRef]
  33. Drucker, D.J. Advances in oral peptide therapeutics. Nat. Rev. Drug Discov. 2020, 19, 277–289. [Google Scholar] [CrossRef]
  34. Ahren, B. DPP-4 Inhibition and the Path to Clinical Proof. Front. Endocrinol. 2019, 10, 376. [Google Scholar] [CrossRef]
  35. Syed, Y.Y. Tirzepatide: First Approval. Drugs 2022, 82, 1213–1220. [Google Scholar] [CrossRef]
  36. Chavda, V.P.; Ajabiya, J.; Teli, D.; Bojarska, J.; Apostolopoulos, V. Tirzepatide, a New Era of Dual-Targeted Treatment for Diabetes and Obesity: A Mini-Review. Molecules 2022, 27, 4315. [Google Scholar] [CrossRef]
  37. Hallakou-Bozec, S.; Vial, G.; Kergoat, M.; Fouqueray, P.; Bolze, S.; Borel, A.L.; Fontaine, E.; Moller, D.E. Mechanism of action of Imeglimin: A novel therapeutic agent for type 2 diabetes. Diabetes Obes. Metab. 2021, 23, 664–673. [Google Scholar] [CrossRef]
  38. DeFronzo, R.A. Chiglitazar: A novel pan-PPAR agonist. Sci. Bull. 2021, 66, 1497–1498. [Google Scholar] [CrossRef]
  39. Xu, H.-R.; Zhang, J.-W.; Chen, W.-L.; Ning, Z.-Q.; Li, X.-N. Pharmacokinetics, Safety and Tolerability of Chiglitazar, A Novel Peroxisome Proliferator-Activated Receptor (PPAR) Pan-Agonist, in Healthy Chinese Volunteers: A Phase I Study. Clin. Drug Investig. 2019, 39, 553–563. [Google Scholar] [CrossRef]
  40. Yan, J.; Zhao, J.; Yang, R.; Zhao, W. Bioactive peptides with antidiabetic properties: A review. Int. J. Food Sci. Technol. 2019, 54, 1909–1919. [Google Scholar] [CrossRef] [Green Version]
  41. Food and Agriculture Organization of the United Nations. Definition and Classification of Commodities: Pulses and Derived Products. 1994. Available online: http://www.fao.org/es/faodef/fdef04e.htm (accessed on 2 June 2015).
  42. Singhal, P.; Kaushik, G.; Mathur, P. Antidiabetic potential of commonly consumed legumes: A review. Crit. Rev. Food Sci. Nutr. 2014, 54, 655–672. [Google Scholar] [CrossRef]
  43. Kamran, F.; Reddy, N. Bioactive peptides from legumes: Functional and nutraceutical potential. Recent Adv. Food Sci. 2018, 1, 134–149. [Google Scholar]
  44. Jakubczyk, A.; Karas, M.; Rybczynska-Tkaczyk, K.; Zielinska, E.; Zielinski, D. Current Trends of Bioactive Peptides-New Sources and Therapeutic Effect. Foods 2020, 9, 846. [Google Scholar] [CrossRef]
  45. Baldeon, M.E.; Felix, C.; Fornasini, M.; Zertuche, F.; Largo, C.; Paucar, M.J.; Ponce, L.; Rangarajan, S.; Yusuf, S.; Lopez-Jaramillo, P. Prevalence of metabolic syndrome and diabetes mellitus type-2 and their association with intake of dairy and legume in Andean communities of Ecuador. PLoS ONE 2021, 16, e0254812. [Google Scholar] [CrossRef]
  46. Rizzello, C.G.; Tagliazucchi, D.; Babini, E.; Sefora Rutella, G.; Taneyo Saa, D.L.; Gianotti, A. Bioactive peptides from vegetable food matrices: Research trends and novel biotechnologies for synthesis and recovery. J. Funct. Foods 2016, 27, 549–569. [Google Scholar] [CrossRef]
  47. Becerra-Tomas, N.; Diaz-Lopez, A.; Rosique-Esteban, N.; Ros, E.; Buil-Cosiales, P.; Corella, D.; Estruch, R.; Fito, M.; Serra-Majem, L.; Aros, F.; et al. Legume consumption is inversely associated with type 2 diabetes incidence in adults: A prospective assessment from the PREDIMED study. Clin. Nutr. 2018, 37, 906–913. [Google Scholar] [CrossRef]
  48. Villegas, R.; Gao, Y.T.; Yang, G.; Li, H.L.; Elasy, T.A.; Zheng, W.; Shu, X.O. Legume and soy food intake and the incidence of type 2 diabetes in the Shanghai Women’s Health Study. Am. J. Clin. Nutr. 2008, 87, 162–167. [Google Scholar] [CrossRef]
  49. Tang, J.; Wan, Y.; Zhao, M.; Zhong, H.; Zheng, J.S.; Feng, F. Legume and soy intake and risk of type 2 diabetes: A systematic review and meta-analysis of prospective cohort studies. Am. J. Clin. Nutr. 2020, 111, 677–688. [Google Scholar] [CrossRef]
  50. Konishi, K.; Wada, K.; Yamakawa, M.; Goto, Y.; Mizuta, F.; Koda, S.; Uji, T.; Tsuji, M.; Nagata, C. Dietary Soy Intake Is Inversely Associated with Risk of Type 2 Diabetes in Japanese Women but Not in Men. J. Nutr. 2019, 149, 1208–1214. [Google Scholar] [CrossRef]
  51. Chelliah, R.; Wei, S.; Daliri, E.B.; Elahi, F.; Yeon, S.J.; Tyagi, A.; Liu, S.; Madar, I.H.; Sultan, G.; Oh, D.H. The Role of Bioactive Peptides in Diabetes and Obesity. Foods 2021, 10, 2220. [Google Scholar] [CrossRef]
  52. Marya; Khan, H.; Nabavi, S.M.; Habtemariam, S. Anti-diabetic potential of peptides: Future prospects as therapeutic agents. Life Sci. 2018, 193, 153–158. [Google Scholar] [CrossRef]
  53. Rivero-Pino, F.; Espejo-Carpio, F.J.; Guadix, E.M. Antidiabetic Food-Derived Peptides for Functional Feeding: Production, Functionality and In Vivo Evidences. Foods 2020, 9, 983. [Google Scholar] [CrossRef]
  54. Bertoglio, J.C.; Calvo, M.A.; Hancke, J.L.; Burgos, R.A.; Riva, A.; Morazzoni, P.; Ponzone, C.; Magni, C.; Duranti, M. Hypoglycemic effect of lupin seed gamma-conglutin in experimental animals and healthy human subjects. Fitoterapia 2011, 82, 933–938. [Google Scholar] [CrossRef]
  55. Méric, E.; Lemieux, S.; Turgeon, S.L.; Bazinet, L. Insulin and glucose responses after ingestion of different loads and forms of vegetable or animal proteins in protein enriched fruit beverages. J. Funct. Foods 2014, 10, 95–103. [Google Scholar] [CrossRef]
  56. Lu, J.; Zeng, Y.; Hou, W.; Zhang, S.; Li, L.; Luo, X.; Xi, W.; Chen, Z.; Xiang, M. The soybean peptide aglycin regulates glucose homeostasis in type 2 diabetic mice via IR/IRS1 pathway. J. Nutr. Biochem. 2012, 23, 1449–1457. [Google Scholar] [CrossRef]
  57. Jiang, H.; Feng, J.; Du, Z.; Zhen, H.; Lin, M.; Jia, S.; Li, T.; Huang, X.; Ostenson, C.G.; Chen, Z. Oral administration of soybean peptide Vglycin normalizes fasting glucose and restores impaired pancreatic function in Type 2 diabetic Wistar rats. J. Nutr. Biochem. 2014, 25, 954–963. [Google Scholar] [CrossRef]
  58. Yamada, Y.; Muraki, A.; Oie, M.; Kanegawa, N.; Oda, A.; Sawashi, Y.; Kaneko, K.; Yoshikawa, M.; Goto, T.; Takahashi, N.; et al. Soymorphin-5, a soy-derived mu-opioid peptide, decreases glucose and triglyceride levels through activating adiponectin and PPARalpha systems in diabetic KKAy mice. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E433–E440. [Google Scholar] [CrossRef] [Green Version]
  59. Lovati, M.R.; Manzoni, C.; Castiglioni, S.; Parolari, A.; Magni, C.; Duranti, M. Lupin seed gamma-conglutin lowers blood glucose in hyperglycaemic rats and increases glucose consumption of HepG2 cells. Br. J. Nutr. 2012, 107, 67–73. [Google Scholar] [CrossRef] [Green Version]
  60. Mojica, L.; Gonzalez de Mejia, E.; Granados-Silvestre, M.Á.; Menjivar, M. Evaluation of the hypoglycemic potential of a black bean hydrolyzed protein isolate and its pure peptides using in silico, in vitro and in vivo approaches. J. Funct. Foods 2017, 31, 274–286. [Google Scholar] [CrossRef]
  61. Gonzalez-Montoya, M.; Hernandez-Ledesma, B.; Mora-Escobedo, R.; Martinez-Villaluenga, C. Bioactive Peptides from Germinated Soybean with Anti-Diabetic Potential by Inhibition of Dipeptidyl Peptidase-IV, alpha-Amylase, and alpha-Glucosidase Enzymes. Int. J. Mol. Sci. 2018, 19, 2883. [Google Scholar] [CrossRef] [Green Version]
  62. Jiang, M.; Yan, H.; He, R.; Ma, Y. Purification and a molecular docking study of α-glucosidase-inhibitory peptides from a soybean protein hydrolysate with ultrasonic pretreatment. Eur. Food Res. Technol. 2018, 244, 1995–2005. [Google Scholar] [CrossRef]
  63. Lammi, C.; Zanoni, C.; Arnoldi, A. Three Peptides from Soy Glycinin Modulate Glucose Metabolism in Human Hepatic HepG2 Cells. Int. J. Mol. Sci. 2015, 16, 27362–27370. [Google Scholar] [CrossRef] [Green Version]
  64. Muñoz, E.B.; Luna-Vital, D.A.; Fornasini, M.; Baldeón, M.E.; Gonzalez de Mejia, E. Gamma-conglutin peptides from Andean lupin legume (Lupinus mutabilis Sweet) enhanced glucose uptake and reduced gluconeogenesis in vitro. J. Funct. Foods 2018, 45, 339–347. [Google Scholar] [CrossRef]
  65. Kwon, D.Y.; Hong, S.M.; Ahn, I.S.; Kim, M.J.; Yang, H.J.; Park, S. Isoflavonoids and peptides from meju, long-term fermented soybeans, increase insulin sensitivity and exert insulinotropic effects in vitro. Nutrition 2011, 27, 244–252. [Google Scholar] [CrossRef]
  66. Oseguera-Toledo, M.E.; Gonzalez de Mejia, E.; Amaya-Llano, S.L. Hard-to-cook bean (Phaseolus vulgaris L.) proteins hydrolyzed by alcalase and bromelain produced bioactive peptide fractions that inhibit targets of type-2 diabetes and oxidative stress. Food Res. Int. 2015, 76, 839–851. [Google Scholar] [CrossRef]
  67. Barnes, M.J.; Uruakpa, F.O.; Udenigwe, C.C. Influence of Cowpea (Vigna unguiculata) Peptides on Insulin Resistance. J. Nutrit. Health Food Sci. 2015, 3, 1–3. [Google Scholar]
  68. Davis, J.; Higginbotham, A.; O’Connor, T.; Moustaid-Moussa, N.; Tebbe, A.; Kim, Y.C.; Cho, K.W.; Shay, N.; Adler, S.; Peterson, R.; et al. Soy protein and isoflavones influence adiposity and development of metabolic syndrome in the obese male ZDF rat. Ann. Nutr. Metab. 2007, 51, 42–52. [Google Scholar] [CrossRef]
  69. Nordentoft, I.; Jeppesen, P.B.; Hong, J.; Abudula, R.; Hermansen, K. Increased Insulin Sensitivity and Changes in the Expression Profile of Key Insulin Regulatory Genes and Beta Cell Transcription Factors in Diabetic KKAy-Mice after Feeding with a Soy Bean Protein Rich Diet High in Isoflavone Content. J. Agric. Food Chem. 2008, 56, 4377–4385. [Google Scholar] [CrossRef]
  70. Gonzalez-Santiago, A.E.; Vargas-Guerrero, B.; Garcia-Lopez, P.M.; Martinez-Ayala, A.L.; Dominguez-Rosales, J.A.; Gurrola-Diaz, C.M. Lupinus albus Conglutin Gamma Modifies the Gene Expressions of Enzymes Involved in Glucose Hepatic Production In Vivo. Plant Foods Hum. Nutr. 2017, 72, 134–140. [Google Scholar] [CrossRef]
  71. Magni, C.; Sessa, F.; Accardo, E.; Vanoni, M.; Morazzoni, P.; Scarafoni, A.; Duranti, M. Conglutin gamma, a lupin seed protein, binds insulin in vitro and reduces plasma glucose levels of hyperglycemic rats. J. Nutr. Biochem. 2004, 15, 646–650. [Google Scholar] [CrossRef]
  72. Vargas-Guerrero, B.; Garcia-Lopez, P.M.; Martinez-Ayala, A.L.; Dominguez-Rosales, J.A.; Gurrola-Diaz, C.M. Administration of Lupinus albus gamma conglutin (Cgamma) to n5 STZ rats augmented Ins-1 gene expression and pancreatic insulin content. Plant Foods Hum. Nutr. 2014, 69, 241–247. [Google Scholar] [CrossRef]
  73. Terruzzi, I.; Senesi, P.; Magni, C.; Montesano, A.; Scarafoni, A.; Luzi, L.; Duranti, M. Insulin-mimetic action of conglutin-gamma, a lupin seed protein, in mouse myoblasts. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 197–205. [Google Scholar] [CrossRef]
  74. Wei, Y.; Zhang, R.; Fang, L.; Qin, X.; Cai, M.; Gu, R.; Lu, J.; Wang, Y. Hypoglycemic effects and biochemical mechanisms of Pea oligopeptide on high-fat diet and streptozotocin-induced diabetic mice. J. Food Biochem. 2019, 43, e13055. [Google Scholar] [CrossRef]
  75. Valencia-Mejia, E.; Batista, K.A.; Fernandez, J.J.A.; Fernandes, K.F. Antihyperglycemic and hypoglycemic activity of naturally occurring peptides and protein hydrolysates from easy-to-cook and hard-to-cook beans (Phaseolus vulgaris L.). Food Res. Int. 2019, 121, 238–246. [Google Scholar] [CrossRef]
  76. de Souza Rocha, T.; Hernandez, L.M.R.; Mojica, L.; Johnson, M.H.; Chang, Y.K.; González de Mejía, E. Germination of Phaseolus vulgaris and alcalase hydrolysis of its proteins produced bioactive peptides capable of improving markers related to type-2 diabetes in vitro. Food Res. Int. 2015, 76, 150–159. [Google Scholar] [CrossRef] [Green Version]
  77. Dun, X.P.; Wang, J.H.; Chen, L.; Lu, J.; Li, F.F.; Zhao, Y.Y.; Cederlund, E.; Bryzgalova, G.; Efendic, S.; Jornvall, H.; et al. Activity of the plant peptide aglycin in mammalian systems. FEBS J. 2007, 274, 751–759. [Google Scholar] [CrossRef]
  78. Hirano, K.; Hirano, H. Interaction of a 43-kDa Receptor-like Protein with a 4-kDa Hormone-like Peptide in Soybean. Biochemistry 2004, 43, 12105–12112. [Google Scholar]
  79. Watanbe, Y. A peptide that stimulates phosphorylation of the plant insulin-binding protein Isolation, primary structure and cDNA cloning. Eur. J. Biochem. 1994, 224, 167–172. [Google Scholar] [CrossRef]
  80. Gressent, F.; Duport, G.; Rahioui, I.; Pauchet, Y.; Bolland, P.; Specty, O.; Rahbe, Y. Biological activity and binding site characteristics of the PA1b Entomotoxin on insects from different orders. J. Insect. Sci. 2007, 7, 12. [Google Scholar] [CrossRef]
  81. Domoney, C. Inhibitors of legume seeds. In Seed Proteins; Shewry, P.R., Casey, R., Eds.; Springer: Dordrecht, The Netherlands, 1999; pp. 635–655. [Google Scholar]
  82. Mane, S.P.; Johnson, S.K.; Duranti, M.; Pareek, V.K.; Utikar, R.P. Lupin seed γ-conglutin: Extraction and purification methods—A review. Trends Food Sci. Technol. 2018, 73, 1–11. [Google Scholar] [CrossRef]
  83. Lima-Cabello, E.; Robles-Bolivar, P.; Alché, J.D.; Jimenez-Lopez, J.C. Narrow Leafed Lupin Beta-Conglutin Proteins Epitopes Identification and Molecular Features Analysis Involved in Cross-Allergenicity to Peanut and Other Legumes. GCB 2016, 2, e29. [Google Scholar] [CrossRef] [Green Version]
  84. Jessica Capraro, A.C.; Luis, A.R.; Chiara, M.; Alessio, S.; Marcello, D. Assessment of the lupin seed glucose-lowering protein intestinal absorption by using in vitro and ex vivo models. Food Chem. 2011, 125, 1279–1283. [Google Scholar] [CrossRef]
  85. Claessens, M.; Saris, W.H.; Van Baak, M.A. Glucagon and insulin responses after ingestion of different amounts of intact and hydrolysed proteins. Br. J. Nutr. 2008, 100, 61–69. [Google Scholar] [CrossRef] [Green Version]
  86. Sánchez, A.; Vázquez, A. Bioactive peptides: A review. FQS 2017, 1, 29–46. [Google Scholar] [CrossRef]
  87. Daliri, E.B.; Oh, D.H.; Lee, B.H. Bioactive Peptides. Foods 2017, 6, 32. [Google Scholar] [CrossRef]
  88. Agyei, D. Bioactive Proteins and Peptides from Soybeans. Recent Pat. Food Nutr. Agric. 2015, 7, 100–107. [Google Scholar] [CrossRef]
  89. Chatterjee, C.; Gleddie, S.; Xiao, C.W. Soybean Bioactive Peptides and Their Functional Properties. Nutrients 2018, 10, 1211. [Google Scholar] [CrossRef] [Green Version]
  90. Shahidi, F.; Zhong, Y. Bioactive Peptides. J. Aoac. Int. 2008, 9, 914–931. [Google Scholar] [CrossRef] [Green Version]
  91. Lammi, C.; Bollati, C.; Ferruzza, S.; Ranaldi, G.; Sambuy, Y.; Arnoldi, A. Soybean- and Lupin-Derived Peptides Inhibit DPP-IV Activity on In Situ Human Intestinal Caco-2 Cells and Ex Vivo Human Serum. Nutrients 2018, 10, 1082. [Google Scholar] [CrossRef] [Green Version]
  92. Ngoh, Y.Y.; Tye, G.J.; Gan, C.Y. The investigation of α-amylase inhibitory activity of selected Pinto bean peptides via preclinical study using AR42J cell. J. Funct. Foods 2017, 35, 641–647. [Google Scholar] [CrossRef]
  93. Ngoh, Y.Y.; Gan, C.Y. Identification of Pinto bean peptides with inhibitory effects on alpha-amylase and angiotensin converting enzyme (ACE) activities using an integrated bioinformatics-assisted approach. Food Chem. 2018, 267, 124–131. [Google Scholar] [CrossRef]
  94. Jakubczyk, A.; Karas, M.; Zlotek, U.; Szymanowska, U. Identification of potential inhibitory peptides of enzymes involved in the metabolic syndrome obtained by simulated gastrointestinal digestion of fermented bean (Phaseolus vulgaris L.) seeds. Food Res. Int. 2017, 100, 489–496. [Google Scholar] [CrossRef]
  95. Mojica, L.; de Mejia, E.G. Optimization of enzymatic production of anti-diabetic peptides from black bean (Phaseolus vulgaris L.) proteins, their characterization and biological potential. Food Funct. 2016, 7, 713–727. [Google Scholar] [CrossRef]
  96. Huang, M.H.; Lin, J.L.; Wang, M.; Ma, Y.; Wang, J.F. High-level expression and activity determination of hypoglycemic peptide aglycin. Mod. Food Sci. Technol. 2020, 36, 143–149. [Google Scholar] [CrossRef]
  97. Wang, R.; Zhao, H.; Pan, X.; Orfila, C.; Lu, W.; Ma, Y. Preparation of bioactive peptides with antidiabetic, antihypertensive, and antioxidant activities and identification of alpha-glucosidase inhibitory peptides from soy protein. Food Sci. Nutr. 2019, 7, 1848–1856. [Google Scholar] [CrossRef]
  98. Mojica, L.; Luna-Vital, D.A.; Gonzalez de Mejia, E. Characterization of peptides from common bean protein isolates and their potential to inhibit markers of type-2 diabetes, hypertension and oxidative stress. J. Sci. Food Agric. 2017, 97, 2401–2410. [Google Scholar] [CrossRef]
  99. Chandrasekaran, S.; Luna-Vital, D.; de Mejia, E.G. Identification and Comparison of Peptides from Chickpea Protein Hydrolysates Using Either Bromelain or Gastrointestinal Enzymes and Their Relationship with Markers of Type 2 Diabetes and Bitterness. Nutrients 2020, 12, 3843. [Google Scholar] [CrossRef]
  100. Sato, K.; Miyasaka, S.; Tsuji, A.; Tachi, H. Isolation and characterization of peptides with dipeptidyl peptidase IV (DPPIV) inhibitory activity from natto using DPPIV from Aspergillus oryzae. Food Chem. 2018, 261, 51–56. [Google Scholar] [CrossRef]
  101. Mojica, L.; Chen, K.; de Mejia, E.G. Impact of commercial precooking of common bean (Phaseolus vulgaris) on the generation of peptides, after pepsin-pancreatin hydrolysis, capable to inhibit dipeptidyl peptidase-IV. J. Food. Sci. 2015, 80, H188–H198. [Google Scholar] [CrossRef]
  102. Mune Mune, M.A.; Minka, S.R.; Henle, T. Investigation on antioxidant, angiotensin converting enzyme and dipeptidyl peptidase IV inhibitory activity of Bambara bean protein hydrolysates. Food Chem. 2018, 250, 162–169. [Google Scholar] [CrossRef]
  103. de Souza Rocha, T.; Hernandez, L.M.R.; Chang, Y.K.; de Mejia, E.G. Impact of germination and enzymatic hydrolysis of cowpea bean (Vigna unguiculata) on the generation of peptides capable of inhibiting dipeptidyl peptidase IV. Food Res. Int. 2014, 64, 799–809. [Google Scholar] [CrossRef]
  104. Castañeda-Pérez, E.; Jiménez-Morales, K.; Quintal-Novelo, C.; Moo-Puc, R.; Chel-Guerrero, L.; Betancur-Ancona, D. Enzymatic protein hydrolysates and ultrafiltered peptide fractions from Cowpea Vigna unguiculata L bean with in vitro antidiabetic potential. J. Iran. Chem. Soc. 2019, 16, 1773–1781. [Google Scholar] [CrossRef]
  105. Cnop, M.; Welsh, N.; Jonas, J.C.; Jörns, A.; Lenzen, S.; Eizirik, D.L. Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: Many differences, few similarities. Diabetes 2005, 54, S97–S107. [Google Scholar] [CrossRef] [Green Version]
  106. Guo, S.D. Insulin signaling, resistance, and the metabolic syndrome: Insights from mouse models into disease mechanisms. J. Endocrinol. 2014, 220, T1–T23. [Google Scholar] [CrossRef] [Green Version]
  107. Newsholme, P.; Bender, K.; Kiely, A.; Brennan, L. Amino acid metabolism, insulin secretion and diabetes. Biochem. Soc. Trans. 2007, 35, 1180–1186. [Google Scholar] [CrossRef]
  108. Hilder, T.L.; Baer, L.A.; Fuller, P.M.; Fuller, C.A.; Grindeland, R.E.; Wade, C.E.; Graves, L.M. Insulin-independent pathways mediating glucose uptake in hindlimb-suspended skeletal muscle. J. Appl. Physiol. 2005, 99, 2181–2188. [Google Scholar] [CrossRef]
  109. Huang, S.; Czech, M.P. The GLUT4 glucose transporter. Cell Metab. 2007, 5, 237–252. [Google Scholar] [CrossRef] [Green Version]
  110. Kwon, D.Y.; Daily, J.W., 3rd; Kim, H.J.; Park, S. Antidiabetic effects of fermented soybean products on type 2 diabetes. Nutr. Res. 2010, 30, 1–13. [Google Scholar] [CrossRef]
  111. Ngoh, Y.Y.; Gan, C.Y. Enzyme-assisted extraction and identification of antioxidative and alpha-amylase inhibitory peptides from Pinto beans (Phaseolus vulgaris cv. Pinto). Food Chem. 2016, 190, 331–337. [Google Scholar] [CrossRef]
  112. Bompard-Gilles, C.; Rousseau, P.; Rougé, P.; Payan, F. Substrate mimicry in the active center of a mammalian α amylase: Structural analysis of an enzyme–inhibitor complex. Structure 1996, 4, 1441–1452. [Google Scholar] [CrossRef] [Green Version]
  113. Nahoum, V.; Farisei, F.; Le-Berre-Anton, V.; Egloff, M.P.; Rouge, P.; Poerio, E.; Payan, F. A plant-seed inhibitor of two classes of alpha-amylases: X-ray analysis of Tenebrio molitor larvae alpha-amylase in complex with the bean Phaseolus vulgaris inhibitor. Acta Crystallogr. Sect. D Struct. Biol. 1999, 55, 360–362. [Google Scholar] [CrossRef]
  114. Patil, P.; Mandal, S.; Tomar, S.K.; Anand, S. Food protein-derived bioactive peptides in management of type 2 diabetes. Eur. J. Nutr. 2015, 54, 863–880. [Google Scholar] [CrossRef]
  115. Arulmozhiraja, S.; Matsuo, N.; Ishitsubo, E.; Okazaki, S.; Shimano, H.; Tokiwa, H. Comparative Binding Analysis of Dipeptidyl Peptidase IV (DPP-4) with Antidiabetic Drugs—An Ab Initio Fragment Molecular Orbital Study. PLoS ONE 2016, 11, e0166275. [Google Scholar] [CrossRef] [Green Version]
  116. Zizzari, A.T.; Pliatsika, D.; Gall, F.M.; Fischer, T.; Riedl, R. New perspectives in oral peptide delivery. Drug Discov. Today 2021, 26, 1097–1105. [Google Scholar] [CrossRef]
  117. Li, Y.; Zhang, W.; Zhao, R.; Zhang, X. Advances in oral peptide drug nanoparticles for diabetes mellitus treatment. Bioact. Mater. 2022, 15, 392–408. [Google Scholar] [CrossRef]
  118. Yap, P.G.; Gan, C.Y. In vivo challenges of anti-diabetic peptide therapeutics: Gastrointestinal stability, toxicity and allergenicity. Trends Food Sci. Technol. 2020, 105, 161–175. [Google Scholar] [CrossRef]
  119. Oseguera-Toledo, M.E. Proteins and bioactive peptides: Mechanism of action in diabetes management. Nutrafoods 2014, 13, 147–157. [Google Scholar] [CrossRef]
  120. Rosenstock, J.; Allison, D.; Birkenfeld, A.L.; Blicher, T.M.; Deenadayalan, S.; Jacobsen, J.B.; Serusclat, P.; Violante, R.; Watada, H.; Davies, M.; et al. Effect of Additional Oral Semaglutide vs Sitagliptin on Glycated Hemoglobin in Adults with Type 2 Diabetes Uncontrolled with Metformin Alone or with Sulfonylurea: The PIONEER 3 Randomized Clinical Trial. JAMA 2019, 321, 1466–1480. [Google Scholar] [CrossRef] [Green Version]
  121. Davies, M.; Pieber, T.R.; Hartoft-Nielsen, M.L.; Hansen, O.K.H.; Jabbour, S.; Rosenstock, J. Effect of Oral Semaglutide Compared with Placebo and Subcutaneous Semaglutide on Glycemic Control in Patients with Type 2 Diabetes: A Randomized Clinical Trial. JAMA 2017, 318, 1460–1470. [Google Scholar] [CrossRef]
  122. Poudwal, S.; Misra, A.; Shende, P. Role of lipid nanocarriers for enhancing oral absorption and bioavailability of insulin and GLP-1 receptor agonists. J. Drug Target. 2021, 29, 834–847. [Google Scholar] [CrossRef]
Figure 1. Mechanism of anti-diabetic effects of 10 representative legume-derived peptides. The amino acid sequence or molecular structure of the peptides are presented in the (right) panel. The anti-diabetic mechanisms of these peptides in corresponding organs are showed in the (left) panel. ↑ means a stimulative effect and ↓ means an inhibitory effect in the text. Peptide 1 works in pancreas; peptides 1 to 5 work in liver; peptide 6 works in muscle and adipose cells; and peptides 7–10 work in intestinal/colon tissue.
Figure 1. Mechanism of anti-diabetic effects of 10 representative legume-derived peptides. The amino acid sequence or molecular structure of the peptides are presented in the (right) panel. The anti-diabetic mechanisms of these peptides in corresponding organs are showed in the (left) panel. ↑ means a stimulative effect and ↓ means an inhibitory effect in the text. Peptide 1 works in pancreas; peptides 1 to 5 work in liver; peptide 6 works in muscle and adipose cells; and peptides 7–10 work in intestinal/colon tissue.
Nutrients 15 01096 g001
Table 1. Clinical drugs in T2D and their target organs/tissues.
Table 1. Clinical drugs in T2D and their target organs/tissues.
Target Organs/TissuesMechanismDrug ClassDrug NameAdverse ReactionDrug Delivery
Pancreatic β-cellβ-cell mass restore, increasing insulin secretionSulfonylureaGlibenclamide, glipizide, glimepiride, gliclazideCause hypoglycemiaOral
MeglitinideRepaglinide, nateglinideNot cause hypoglycemiaOral
Pancreatic α-cellIncreasing glucagon secretionGLP-1 agonistExenatide, lixisenatide, liraglutide, dulaglutide, exenatide extend-release, semaglutide, albiglutideGastrointestinal and hypoglycemic reactionsInject/Oral
Amylin analogPramlintideCause hypoglycemicInject
BiguanideMetforminLactic acidemia and ketouriaOral
LiverDecreasing hepatic glucose productionThiazolidinedionePioglitazone, rosiglitazoneWeight gain, edema, cardiac problemsOral
InsulinRapid-, short-, intermediate-, long-acting insulinHypoglycemia and edemaInject
BiguanideMetforminLactic acidemia and ketouriaOral
Muscle and adipose tissueIncreasing glucose uptake and glycolysisBiguanideMetforminLactic acidemia and ketouriaOral
ThiazolidinedionePioglitazone, rosiglitazoneWeight gain, edema, cardiac problemsOral
InsulinRapid-, short-, intermediate-, long-acting insulinHypoglycemia and edemaInject
α-glucosidase inhibitorAcarbose, miglitol, vogliboseGastrointestinal reactionOral
IntestineIncreasing incretin activity, enhancing glucose absorptionDPP-4 inhibitorSitagliptin, saxagliptin, linagliptin, vidaglipti, alogliptin, teneligliptin, gemigliptinMinor adverse drug reactionsOral
GLP-1 agonistExenatide, lixisenatide, liraglutide, dulaglutide, exenatide extend-release, semaglutide, albiglutideGastrointestinal and hypoglycemic reactionsInject/Oral
BiguanideMetforminLactic acidemia and ketouriaOral
ColonBalancing gut microbiotaGLP-1 agonistExenatide, lixisenatide, liraglutide, dulaglutide, exenatide extend-release, semaglutide, albiglutideGastrointestinal and hypoglycemic reactionsInject/Oral
KidneyInhibiting glucose reabsorptionSGLT-2 inhibitorDapagliflozin, canagliflozin, empagliflozin, ertugliflozin, SotagliflozinUrinary genital infection risk, ketoacidosis riskOral
Central nervous systemLowing neurotransmittersDopamine-receptor agonistBromocriptineGastrointestinal reactionOral
Table 2. Risk of T2D in relation to the average intake of legumes and their proteins.
Table 2. Risk of T2D in relation to the average intake of legumes and their proteins.
SourceConsumption(g/d)People TestedHRs or RR (95% CIs)Reference
Lentils6.6Caucasian0.67 (0.46, 0.98)[47]
Chickpea5.0Caucasian0.68 (0.46, 1.00)[47]
Soybean32.0Chinese women0.57 (0.48, 0.60)[48]
Soy protein11.0−15.0Chinese/Japanese0.84 (0.75, 0.95)[49]
Soy protein13.6Japanese women0.46 (0.30, 0.70)[50]
CIs, confidence intervals; HRs, hazard ratios; RR, relative risk.
Table 3. Hypoglycemic effect and mechanism of legume peptides and hydrolysates in vivo and in vitro.
Table 3. Hypoglycemic effect and mechanism of legume peptides and hydrolysates in vivo and in vitro.
PeptideAmino Acid SequenceSourceModelFeeding/Treat PatternHypoglycemic Effect and IndexMechanismReference
AglycinASCNGVCSPFEMPPCGSSACRCIPVGLVVGYCRHPSGSoybeanSTZ/HFD-induced diabetic BALB/c mice, C2C12 cell50 mg/kg/d, 4 weeksBG↓, OGTT↑, insulin tolerance↑, p-IR↑, p-IRS1↑, p-Akt↑, GLUT4↑, glucose uptake↑Increasing insulin receptor via IR/IRS1 pathway, enhancing glucose uptake[56]
VglycinVSCNGVCSPFEMPPCGSSACRCIPYGLVVGNCRHPSGPea seed/germinating pea seedSTZ/HFD-induced diabetic rats, HepG2 cell, L02 cell4 weeksBW↓, food intake↓, FPG↑, PARP↑, PDX1↑, GSK3α/β↑, GLUT4↑, p85-PI3-kinase↓, p-Akt↓Impairing glucose tolerance, restoring pancreatic function, enhancing insulin signaling by activating the IR/Akt pathway[57]
T1D SD rat, STZ-induced T2D C57BL/6 mice, INS-1 832/13 cell80 mg/kg/d, 4 weeks, 8 weeksBW↓, lee’s index↓, food intake↓, FPG↓, BG↓, ITT↑, pancreatic islet↑, plasma insulin↑, glucose-stimulated glucagon↓, pancreatic mass↑, β-cell area and mass↑, Ki67/PCNA immunostaining area↑, TUNEL-positive/total β-cells↓, Erk↑Promoting the proliferation of β-cells via the IR/Akt/Erk pathway[14]
HFD-C57BL/6J mice, HepG2 cell15 weeksBW↓, FPG↓, glucose tolerance↑, ITT↑, FA β-oxidation↑, FAS↓Improving insulin sensitivity and glucose tolerance, enhancing β-oxidation and inhibiting FAS via AMPK pathway and down-regulating the FAS[15]
Soymorphin-5YPFVVSoybeanKKAy mice10 mg/kg/d, 5 weeksBG↓, plasma insulin↓, TG↓, adiponectin↑, liver TG↓, Adipor2↑, PPARα↑, AOX1↑, CPT1↑, UCP2 1↑Increasing insulin sensitivity, improving glucose and lipid metabolism via activation of the adiponectin and PPARα system[58]
Soy protein/SoybeanZDF ratSoy protein, 11 weeksBW↓, total adiposity↓, total and liver adiposity↓, glucose level↓, insulin↓, GLUT4↑, PPAR-r↑, FAS↓, GPDH↓Maintenance of peripheral (adipose tissue) insulin signaling[68]
Soy protein/SoybeanKKAy miceHigh content isoflavone soy protein, 9 weeksFPG↓, insulin↓, TG↓, TC↓, GLUT2↑, GLUT3↑, Ins1↑, Ins2↑, IGF1↓, β2/Neurod1↓, cholecystokinin↓, LDLR↑Improving glucose and insulin sensitivity[69]
γ-conglutin/Lupin beanSD rat, HepG2 cell10 μmol/L, 24 or 48 hGlucose consumption↑Maintaining glucose homeostasis[59]
γ-conglutin/Lupin beanSTZ-induced Wistar rat150 mg/kg, 1 weekG6pc↓, Fbp1↓, Pck1↓Regulating glucose metabolism mainly through G6pc inhibition[70]
γ-conglutin/Lupin beanGlucose-administrated male rat50, 100 and 200 mg/kg BW, 0, 30, 60 and 90 minBG↓, AUC↓Hypoglycemic effect[54]
γ-conglutin/Lupin beanGlucose-administrated male rat30, 60 and 120 mg/kg BW, 30, 60, 90, and 120 minPlasma glucose↓Hypoglycemic effect[71]
γ-conglutin/Lupin beanSTZ administrated rat120 mg/kg in saline, 1 weekGlucose↓, Ins1↑, pancreatic insulin↑, serum insulin↑Hypoglycemic effect[72]
γ-conglutin/Lupin beanC2C12 cell0.5 mg/mL,72 hPlasma glucose↓, IRS1 protein↑, p85-PI3 kinase↑, Akt1 protein↑, eIF4E protein↑, p70S6 K↑, ERK1↑, ERK2↑, MHC protein↑, myogenin↑Regulating muscle energy metabolism, protein synthesis and MHC gene transcription through insulin signaling pathway[73]
HPI/pure peptideAKSPLF,
ATNPL,
FFEELN,
LSVSVL
Black beanSTZ-induced Wistar rats, Caco-2 cell100, 150 and 200 mg HPI/kg/BW, 15 d, pure peptide (100 µM), HPI (10 mg/mL)BW↓, BG↓, postprandial glucose level↓, insulin↓, GLP-1↑, OGTT↓, glucose absorption↓, GLUT2↓, SGLT1↓Reducing glucose absorption via blocking glucose transporters GLUT2 and SGLT1(in silico)[60]
Synthetic peptideIAVPGEVA,
IAVPTGVA,
LPYP
Soy glycininHepG2 cell50 and 100 µMGlucose uptake↑, Akt↑, GSK3α/β↓, GLUT4↑, GLUT1↑Enhancing glucose uptake through GLUT1 and GLUT4 activation, Akt and AMPK pathway[63]
Hydrolysate/Soy proteinAlloxan-induced male Kunming mice47.5 mg/kg/d, 3 weeksFBG↓Hypoglycemic effect[62]
Hydrolytic oligopeptide/PeaSTZ/HFD-induced diabetic mice800, 1600 and 3200 mg/kg BW, 4 weeksFBG↓, BW↓, OGTT↑, serum insulin↑, TC↓, TG↓, HDL-C↑, fatty acid anion ↓, liver and muscle glycogen↑Enhancing insulin sensitivity[74]
Protein/peptide
fraction
<10 kDaCommon
bean
Male Wistar rat0.5 and 5 mg/kgGlucose level↓, glucose uptake↑Hypoglycemic activity[75]
γ-conglutin hydrolysate/Lupin beanCaco-2 cells, 3T3-L1 cell, HepG2 cell2 and 5 mg/mLGlucose uptake↑, gluconeogenesis↓, PEPCK↓, GLUT4↑, HepG2 glucose production↓Inhibiting DPP-4, improving insulin receptor sensitivity, inhibiting hepatic gluconeogenesis through GLUT4 activation[64]
Peptide/CowpeaRat L6 skeletal muscle cellVarious dosesAkt↑Activating the insulin signaling pathway[67]
Water extract peptide<3 kDaFermented soybean3T3-L1 cell, Min6 cells NCI-H716 cell, Human embryo kidney 293 cell5 µg/mLGlucose uptake↑, triacylglycerol↑, PPAR-r↑, β-cell viability↑, insulin secretion↑, cell proliferation↑, PDX1↑Increasing insulin sensitivity and exerting insulinotropic via PPAR-r activation[65]
Hydrolysate/ fraction<1 and 1–3 kDaHard-to-cook beanINS-1E cell1 g Hydrolysate treatmentIncrease insulin secretion up to 57%Increasing insulin secretion[66]
Hydrolysate/Common
Bean(G0-0h)
INS-1E pancreatic β-cell2 mg SP/mLIncrease insulin secretion 45% from the basal stateIncreasing insulin secretion[76]
G0-0h, the non-germinated and non-hydrolyzed sample; HPI, hydrolyzed protein isolate; SP, soluble protein; ZDF, Zucker diabetic fatty; ↑stimulative effect; ↓inhibitory effect.
Table 4. Legume-derived peptide inhibitors targeting α-amylase, α-glucosidase, and DPP-4.
Table 4. Legume-derived peptide inhibitors targeting α-amylase, α-glucosidase, and DPP-4.
NO.Peptide SequenceMW(DA)SourceTargetIC50, %Reference
1LSSLEMGSLGALFVCM1658.02Pinto beanα-amylase0.31 mM[92]
2PLPLHMLP917.18Pinto beanα-amylase5.92 mM[92]
3PPMHLP690.87Pinto beanα-amylase6.08 mM[92]
4PPHMLP690.81Pinto beanα-amylase23.33 ± 0.15 mM[93]
5PPHMGGP691.81Pinto beanα-amylase6.14 mM[92]
6PLPWGAGF843.98Pinto beanα-amylase6.64 mM[92]
7SPQSPPFATPLW1327.50Chickpeaα-amylase−8.40 kcal/mol[99]
8FVVAEQAGNEEGFE1525.59Fermented bean seedα-amylase0.04−0.65 μg/mL[94]
9SGGGGGGVAGAATASR1232.29Fermented bean seedα-amylase0.59−2.12 μg/mL[94]
10GSGGGGGGGFGGPRR1232.29Fermented bean seedα-amylase0.59−2.12 μg/mL[94]
11INEGSLLLPH1092.26Fermented bean seedα-amylase0.04−0.65 μg/mL[94]
12GGYQGGGYGGNSGGGYGNRG1791.77Fermented bean seedα-amylase0.59−2.12 μg/mL[94]
13GGSGGGGGSSSGRRP1232.24Fermented bean seedα-amylase0.59−2.12 μg/mL[94]
14GDTVTVEFDTFLSR1586.72Fermented bean seedα-amylase0.59−2.12 μg/mL[94]
15NEGEAH655.62Hard-to-cook beanα-amylase−12.84 REU[66]
16FFL425.53Hard-to-cook beanα-amylase−8.22 REU[66]
17WEVM563.67Black beanα-amylase0.04 µM (ki)[95]
18AKSPLF661.79Black beanα-amylase, DPP-40.03 µM (ki), 0.08 µM (ki)[95]
19QQEG460.44Hard-to-cook beanα-amylase, DPP-4−7.03 REU, −7.29 REU[66]
20Aglycin 13743.40Soybean, peaα-glucosidase36.48 μM[96]
21LLPLPVLK892.18Soybeanα-glucosidase237.43 ± 0.52 µM[97]
22SWLRL673.80Soybeanα-glucosidase182.05 ± 0.74 µM[97]
23WLRL586.73Soybeanα-glucosidase162.29 ± 0.74 µM[97]
24GSR318.33Soybeanα-glucosidase20.40 µM[62]
25EAK346.38Soybeanα-glucosidase520.20 µM[62]
26TTGGKGGK704.77Black beanα-glucosidase0.27 µM (ki)[95]
27KKSSG505.57Common beanα-glucosidase, DPP-449.34 ± 6.5%, 0.64 ± 0.16 mg/mL[98]
28GGGLHK567.65Common beanα-glucosidase, DPP-446.10 ± 8.30%, 0.61 ± 0.10 mg/mL[98]
29CPGNK517.60Common beanα-glucosidase, DPP-437.60 ± 6.8%, 0.87 ± 0.02 mg/mL[98]
30KTYGL580.68Common beanα-glucosidase, DPP-436.30 ± 8.80%, 0.03 mg/mL[98]
31YVDGSGTPLT1009.08Chickpeaα-glucosidase, DPP-4−7.30 kcal/mol,
−8.20 kcal/mol
[99]
32IAVPTGVA726.87SoybeanDPP-4223.20 µM (in situ)[91]
33LTFPGSAED935.97Lupin beanDPP-4207.50 µM (in situ)[91]
34EGLELLLLLLAG1253.52Black beanDPP-40.06 µM (ki)[95]
35FEELN650.69Black beanDPP-40.10 µM (ki)[95]
36RGPLVNPDPKPFL1449.72Common beanDPP-414.04 kcal/mol[76]
37KL259.34Fermented soybeanDPP-441.40 ± 2.68 µg/mL[100]
38LR287.35Fermented soybeanDPP-4598.02 ± 18.35 µg/mL[100]
39PHPATSGGGL892.97ChickpeaDPP-4−8.20 kcal/mol[99]
40LLSL444.57Hard-to-cook beanDPP-4−11.75 REU[66]
1 The amino acid sequence of aglycin is in Table 3. Amino acid nomenclature: C, cysteine; H, histidine; I, isoleucine; M, methionine; S, serine; V, valine; A, alanine; G, glycine; L, leucine; P, proline; T, threonine; F, phenylalanine; R, arginine; Y, tyrosine, W, tryptophan; D, aspartic acid; N, asparagine; E, glutamic acid, Q, glutamine; K, lysine. Ki, inhibition constant expressed in µM.
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

Hu, K.; Huang, H.; Li, H.; Wei, Y.; Yao, C. Legume-Derived Bioactive Peptides in Type 2 Diabetes: Opportunities and Challenges. Nutrients 2023, 15, 1096. https://doi.org/10.3390/nu15051096

AMA Style

Hu K, Huang H, Li H, Wei Y, Yao C. Legume-Derived Bioactive Peptides in Type 2 Diabetes: Opportunities and Challenges. Nutrients. 2023; 15(5):1096. https://doi.org/10.3390/nu15051096

Chicago/Turabian Style

Hu, Kanghong, Huizhong Huang, Hanluo Li, Yanhong Wei, and Chenguang Yao. 2023. "Legume-Derived Bioactive Peptides in Type 2 Diabetes: Opportunities and Challenges" Nutrients 15, no. 5: 1096. https://doi.org/10.3390/nu15051096

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