Natural products from plants targeting key enzymes for the future development of antidiabetic agents

R. Mata *, L. Flores-Bocanegra , B. Ovalle-Magallanes and M. Figueroa
Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico. E-mail: rachel@unam.mx

Received 31st January 2023

First published on 7th June 2023


Abstract

Covering: 2000 to January 2023

Diabetes is a metabolic disease of serious concern nowadays, with a negative economic impact. In 2021, the International Diabetes Federation estimated that more than 537 million adults live with diabetes, causing over 6.7 million deaths in that year. Intensive scientific research on medicinal plants in the last 100 years reveals that herbal drugs have been an essential source of products for developing antidiabetic agents acting on different physiological targets. This review summarizes recent research from 2000 to 2022 on plant natural compounds affecting selected crucial enzymes (dipeptidyl peptidase IV, diacylglycerol acyltransferase, fructose 1,6-biphosphatase, glucokinase, and fructokinase) involved in glucose homeostasis. Enzyme-aimed treatments usually induce reversible inhibition, irreversible by covalent changes of the objective enzymes, or bind non-covalently but so tightly that their inhibition is irreversible. Depending on the binding site, these inhibitors could be orthosteric or allosteric; in any case, the desired pharmacological action is achieved. One crucial advantage of targeting enzymes in drug discovery is that the required assays are usually simple, using biochemical experiments capable of analyzing enzyme activity.


image file: d3np00007a-p1.tif

R. Mata

Rachel Mata was born in Caracas, Venezuela, in 1949. She received her B.S. degree in Pharmacy from the Universidad Central de Venezuela in 1971 and her M.S. (1976) and PhD (1979) degrees at Purdue University, IN, USA, under the direction of Dr Jerry L. McLaughlin. Following a postdoctoral stay at the Instituto de Química at Universidad Nacional Autónoma de México (UNAM), she became an Assistant Professor (1984–1987), Associate Professor (1987–1990), Visiting Professor at the University of Rhode Island, TRI, USA (1990–1991), and Professor (1991–2016) at Facultad de Química at UNAM, where she is currently Professor Emerita. Her work has contributed significantly to knowledge of Mexican traditional medicine by establishing the chemical composition and biological properties of many widely used herbal drugs.

image file: d3np00007a-p2.tif

L. Flores-Bocanegra

Laura Flores-Bocanegra received her PhD degree in Chemistry from the Universidad Nacional Autónoma de México (UNAM), and her research topic dealt mainly with the study of medicinal plants from Mexico. From 2018 to 2021, she worked as a Postdoctoral Fellow at the University of North Carolina at Greensboro, NC, USA, as a part of Professor Nicholas H. Oberlies's team. During this period, Dr Flores-Bocanegra worked in the chemical study of the medicinal plant Kratom and several filamentous fungi. She isolated diverse secondary metabolites with different activities such as opioid agonist, cytotoxic, and antibacterial. She became an Assistant Professor at the Facultad de Química at UNAM in 2023. The main research interests of Dr Flores-Bocanegra involve the isolation and characterization of bioactive compounds from medicinal plants as well as fungal species, their pharmacological evaluation, and the validation of analytical methods for the quality control of herbal drugs.

image file: d3np00007a-p3.tif

B. Ovalle-Magallanes

Berenice Ovalle-Magallanes received her PhD degree in Pharmacology at the Facultad de Química (FQ) of the Universidad Nacional Autónoma de México (UNAM) in 2016. After conducting research work at the Department of Pharmacology and Physiology of The Université de Montréal and the Department of Biology of the FQ, UNAM, she became interested in developing and applying in vitro cell-based models to elucidate mechanisms of action of natural products. Since 2019, she has worked as Associate Professor and project group leader under the Young Researchers Program at UNAM. Her research interests include molecular targets in liver and skeletal muscle (using murine and human cells) that regulate glycaemia and cholesterol synthesis and studying potential natural products–drug interactions from edible and medicinal plant species used in Traditional Mexican Cuisine and Medicine.

image file: d3np00007a-p4.tif

M. Figueroa

Mario Figueroa received his B.S. degree in Pharmaceutical and Biological Chemistry in 2004 and his M.S. (2006) and PhD (2009) degrees in Chemistry from the Universidad Nacional Autónoma de México (UNAM) under the mentorship of Professor Rachel Mata. Then, from 2009 to 2013, he worked as a Postdoctoral Fellow at Lehman College, NY, USA, and The University of North Carolina at Greensboro, NC, USA, in the laboratories of Professors Edward J. Kennelly and Nicholas H. Oberlies, respectively. During this period, Dr Figueroa discovered numerous new natural compounds with potential anticancer and anti-MRSA properties from plants and fungi. In 2013, Dr Figueroa returned to UNAM as Associate Professor at the Facultad de Química, and in 2021, he became a Full Professor. His research focuses on the chemistry and biology of fungi and bacteria from unexplored sources in Mexico.


1. Introduction

Diabetes mellitus (DM) is a metabolic disorder characterized by chronic hyperglycemia resulting from insulin secretion and resistance defects. According to the International Diabetes Federation, DM is one of the major leading challenges of the 21st century. In 2021, it was estimated that more than 537 million adults lived with diabetes, causing over 6.7 million deaths that year. The global prevalence of DM is expected to rise to 700 million by 2045. Type II DM (TIIDM) accounts for ∼90% of all cases, and obesity and lifestyle are the major risk factors for developing the disease.1

TIIDM results from insulin resistance and pancreatic β-cells dysfunction, which can lead to micro and macro-vascular complications, often the major causes of mortality among patients.1 Thus, the goal of the treatment of TIIDM is to control blood glucose as measured by glycated hemoglobin, preventing these complications. Usually, the therapy is patient-focused and depends on comorbidities and other factors, but most patients receive combined treatments. The American Diabetes Association and other organizations have designed a few combined-therapy schemes.2 Drug treatments and insulin therapy are prescribed if the higher blood glucose levels cannot be regulated with diet, exercise, and standard drugs.

The most relevant drugs for TIIDM management and their risk/benefit profiles are summarized below:

(i) The biguanide metformin, the first-line oral blood glucose-lowering agent, decreases glucose production in the liver and improves insulin sensitivity, primarily by activating adenosine monophosphate-activated protein kinase (AMPK). Metformin is linked to the traditional herbal medicine Galega officinalis L. (Fabaceae), rich in galegine, another biguanide. It has been widely used to treat the symptoms of diabetes in Europe since the 18th century. The ability of galegine to lower blood glucose was discovered in the 1920s, and after that, some galegine derivatives, including metformin, were prepared, and used to control diabetes during the 1930s. Later, metformin was revived in the search for antimalarial agents in the 1940s and, during clinical tests, proved helpful in lowering blood glucose. In 1994 was approved in the US as an antidiabetic agent.3,4 Metformin possesses a good risk/benefit profile, as it induces minor side effects and has a low chance of provoking hypoglycemia or weight gain. For such characteristics, the American Diabetes Association and the European Association for the Study of Diabetes recommend metformin as initial antihyperglycemic therapy for patients with TIIDM.3,4 Nonetheless, in one in three diabetic patients receiving metformin experiments drowsiness, vomiting, anorexia, diarrhea, nausea, and dyspepsia. These side effects can be overcome using extended-release preparations. Additional precautions should be taken when prescribing elderly patients or with severe renal insufficiency, as metformin can cause vitamin B12, folic acid deficiency, and lactic acidosis. The main problem to overcome when using metformin is its lost of efficacy when TIIDM progresses to a state of low insulin levels, limiting its use in patients with advanced pancreatic β-cells dysfunction.3,4

(ii) Sulfonylureas promote insulin secretion by blocking KATP channels in pancreatic β-cells.3 Second-generation sulfonylureas glipizide, glimepiride, and glibenclamide are prescribed as second-line treatment options for the management of TIIDM. Although these are potent and effective antidiabetic drugs, concerns arise as they provoke hypoglycemia. Also, their combination with other medications, such as beta-adrenergic antagonists, aspirin, allopurinol, fibrates, and other antidiabetic agents like insulin, could increase the risk of hypoglycemic shock. These risks are diminished with a closure monitoring of glycemia and drug–drug interactions. However, typical side effects like weight gain, headaches, dizziness, and nausea provoke treatment abandonment and failure, thus leading to the prescription of another secretagogue medication, such as meglitinides, a class of non-sulfonylurea drugs, or others.3

(iii) Thiazolidinediones improve insulin sensitivity in different body tissues by acting as peroxisome proliferator-activated receptor (PPAR) agonists.3 The prescription of drugs in this class, i.e., troglitazone, rosiglitazone, and pioglitazone, is restricted and limited in most countries due to a poor risk/benefits profile. For instance, the significant side effects of these drugs include weight gain, edema, and hepatic steatosis.3 Although troglitazone was withdrawn from commercialization due to hepatotoxicity and other reports indicate rosiglitazone and pioglitazone induce cardiovascular toxicity and bladder cancer, respectively, recent analyses suggest these safety issues should be addressed again to assess risk/benefit when thiazolidinediones are prescribed.3

(iv) Sodium–glucose cotransporter type 2 (SGLT2) inhibitors, like the gliflozins, which were inspired by phlorizin, a dihydrochalcone glucoside found in many Rosaceae species of the genus Malus Mill, Pyrus L., and Fragaria L. Phlorizin is an inhibitor of SGLT1 and SGLT2 because it competes with D-glucose for binding to the carrier; this action reduces renal glucose transport, lowering the amount of glucose in the blood in an insulin-independent mechanism. As a result, glucose is excreted in the urine.3,5 SGLT2 inhibitors canagliflozin, dapagliflozin, and empagliflozin are well tolerated and have demonstrated their capacity to modestly reduce body weight, blood pressure, and glucotoxicity by antioxidant mechanisms and to improve liver and pancreatic β-cells functioning. Common side effects such as genital mycosis and urinary tract infections, and less frequently, urosepsis and pyelonephritis appear. Therefore, SGLT2 inhibitors are regarded as an excellent alternative to use in combination with other antidiabetic agents, such as dipeptidyl peptidase-IV (DPP-IV) inhibitors.3,5

(v) α-Glucosidase inhibitors hinder the digestion of complex carbohydrates to monosaccharides in the small intestine. The drugs used in therapeutic are natural products or derived from natural products. Thus, acarbose, a pseudo-tetrasaccharide, is a natural microbial product produced from cultures of Actinoplanes utahencis Couch (Micromonosporaceae), which possesses nitrogen between the first and second glucose molecule and has a structure like an oligosaccharide. It was introduced in 1990 for the treatment of TIIDM.6 Voglibose and miglitol are structurally unrelated to glucosidase substrates, possessing piperidine-3,4,5-triol and tetraol-cyclohexane moieties, respectively. Streptomyces hygroscopicus Jensen (Streptomycetaceae) produce valiolamine, the precursor of voglibose. Miglitol is produced from nojirimycin, first reported in 1966 as metabolite of S. roseochromogenes Sugai (Streptomycetaceae), and later from deoxynojirimycin, isolated from the roots of mulberry (Morus Alba L., Moraceae). Mulberry is a traditional Chinese herb with a long history of use.6,7 Furthermore, mulberry leaves are widely used for treating diabetes. Miglitol has the potential as a therapeutic drug against obesity.3,8 Acarbose, miglitol, and voglibose increased glucagon-like peptide-1 (GLP-1), therefore potentiating insulin secretion. Besides reducing postprandial hyperglycemia, acarbose also induces weight loss and lowers total postprandial triglycerides. Significant side effects of α-glucosidase inhibitors are related to gastrointestinal discomforts, such as nausea, abdominal pain, bloating, flatulence, and diarrhea.3

(vi) Incretin therapies (IT), comprise glucagon-like peptide-1 (GLP-1) receptor agonists and dipeptidyl peptidase-IV (DPP-IV) inhibitors. The GLP-1 receptor agonists (e.g., exenatide and liraglutide) stimulate insulin secretion and suppress glucagon synthesis. The first agonist of GLP-1 used in therapeutics was exenatide, a peptide isolated from the saliva of the Gila monster.3 This peptide is a potent antidiabetic agent that reduces body weight combined with metformin. Liraglutide also induces weight loss when given with metformin or a sulfonylurea and decreases glycosylated hemoglobin and systolic pressure. Both drugs induce mild gastrointestinal effects, and GLP-1 analogs, in general, are contraindicated in renal failure. Two significant setbacks in their use are that they must be given in the form of injection and that their cost makes them unavailable for most patients in developing countries.3

GLP-1 acts within 2–4 min and is rapidly inactivated by the DPP-IV enzyme. DPP-IV inhibitors delay the degradation of GLP-1 and the glucose-dependent insulinotropic peptide (GIP). Gliptins are the main class of DPP-IV inhibitors (sitagliptin, saxagliptin, linagliptin, and alogliptin). These drugs reduce glycosylated hemoglobin and have a low risk of hypoglycemia without affecting body weight. GLP-1 receptor agonists and DPP-IV inhibitors improve pancreatic β-cell physiology and reduce apoptosis-associated glucotoxicity. Furthermore, IT improves cardiovascular markers related to dyslipidemia, hypertension, and obesity, reasons for which IT accounts for 30% of antidiabetic drugs in clinical development. In particular, the use of DPP-IV inhibitors has grown significantly during the last decade. Despite its benefits, IT induces side effects such as infections in the respiratory tract, headaches, skin lesions, bladder infections, gastrointestinal problems, and nasopharyngitis, as well as raises concerns about the risk of developing pancreatic cancer. These undesired actions justify the search for new DPP-IV inhibitors.3

(vii) Finally, insulin, alone or in combination with other drugs, is prescribed if proper blood sugar levels are not reached with lifestyle changes and other medications.3 Insulin itself is a natural product. Currently, several insulin formulations possess different pharmacokinetic and pharmacodynamic profiles with high potency and low risks of hypoglycemia. Most prescribed insulin formulations must be given by injection (except for inhaled), which provokes lipohypertrophy due to repeated use of the same area for subcutaneous injection, thus leading to low insulin absorption and deficient glycemic control. Furthermore, insulin formulations induce weight gain, which is detrimental to TIIDM control, formulation excipients are potential allergens, and medication costs have increased exponentially in the last decade.3 These factors lead to treatment abandonment and the appearance of potentially deadly complications, such as diabetic ketoacidosis. Therefore, combination therapies using adequate antidiabetic agents of those mentioned above would contribute to improving insulin therapy.3

Understanding the intricate mechanisms implicated in TIIDM has prompted the search for novel therapeutic agents, including some crucial enzymes for glucose homeostasis. This review will focus on specific enzymes that play a critical role in the pathophysiology of diabetes. The enzymes selected were DPP-IV considering that IT accounts for 30% of antidiabetic drugs in clinical development, and some of its inhibitors (e.g., sitagliptin, saxagliptin, linagliptin, and alogliptin) are now leading drugs for treating diabetes, after insulin and metformin.3 Diacylglycerol acyltransferase (DGAT) is a critical enzyme in triacylglycerol synthesis (TAGS). The intensified activity of DGAT in patients with TIIDM can cause β-cell dysfunction. Therefore, selective DGAT inhibitors might be essential in managing TIIDM and other metabolic diseases. Fructose 1,6-bisphosphatase-1 (FBPase), an enzyme involved in gluconeogenesis, is relevant in glucose metabolism. In TIIDM, gluconeogenesis significantly contributes to an excess of glucose. Reducing this excess would mitigate high glucose concentrations in the blood and tissues. Glucokinase (GK) in the pancreatic β-cells serves as a glucose sensor, increasing insulin secretion as blood glucose rises. Fructokinase or hexokinase (KHK) is an enzyme that converts fructose into fructose 1-phosphate. The activation of the latter enzyme leads to the formation of uric acid, which causes inflammation in the cells of pancreatic islets and, thus, insulin resistance. The inhibition of this pathway could control hyperglycemia. No therapeutic agents affect DAGT, TAGS, FBPase, GK, or KHK, although some compounds targeting these enzymes are now in clinical studies.3 As pointed out in the above paragraphs, natural antidiabetic drugs from plants or other natural sources have inspired the development of some therapeutic agents in the past.9 Therefore, it is highly probable that other natural products targeting the selected enzymes but DPP-IV yield new drugs for treating diabetes.

Finally, it is important to point out that the enzyme-aimed treatments usually induce reversible inhibition, covalent changes of the objective enzymes, or bind non-covalently but so tightly that their inhibition is irreversible. In any case, the desired pharmacological action is achieved. One crucial advantage of targeting enzymes in drug discovery is that the required assays are usually simple, using biochemical experiments capable of analyzing enzyme activity. Enzyme-based drug discovery can also benefit from crystallography analysis of the enzymes and in silico assays.10 The assays must be completed with proper kinetics studies, demonstrating reagent and response stability.

2. Methods

A systematic and comprehensive search for literature was carried out in various scientific databases, including Google Scholar, PubMed, ScienceDirect, Scopus, and Springer Link, to obtain the desired information available in journals and books from 2000–2022. Searching keywords were “dipeptidyl peptidase IV inhibitors”, “diacylglycerol acyltransferase inhibitors”, “fructose 1,6-bisphosphatase inhibitors”, “glucokinase activators”, “hexokinase inhibitors” and “fructokinase inhibitors” over 14[thin space (1/6-em)]000, 680, 470, 680, and 80 hits, respectively, were found. After that, a comprehensive refinement using the keywords “Diabetes mellitus”, “medicinal plants”, “phytochemicals”, and “natural products”, alone and in combination, was performed. Synthetic and in silico studies and activity data from extracts or fractions were excluded. Chemical structures were drawn using ChemBioDraw Ultra 14.0 (Cambridge Software), and protein structures were rendered with PyMOL (Schrödinger).

3. Dipeptidyl peptidase IV inhibitors

3.1. Structure of DPP-IV

DPP-IV is a multifunctional type II glycoprotein of the prolyl oligopeptidase family. The enzyme cuts peptides with penultimate Pro or Ala residues from the N-terminus of at least 21 bioactive peptides.11

The GIP and GLP-1 are the most relevant substrates of DPP-IV concerning TIIDM, but there are other biological ligands. DPP-IV is an integral membrane protein ubiquitously distributed and expressed in intestinal, kidney, vascular endothelium, immune system, liver, and pancreas cells.11

Human DPP-IV is a homodimer with two subunits (PDB code: 4J3J; Fig. 1). Each subunit comprises 766 amino acids (110 kDa), with the β-propeller and the α/β-hydrolase domains surrounding a space containing the active site. Functionally, these two domains are extracellular (amino acids 29–766). The structure has two channels between the hydrolase and propeller domains next to the catalytic site, through which substrates and ligands could contact the active site. DPP-IV also possesses a small tail (intracellular, amino acids 1–6) and a transmembrane region (amino acids 7–28). The eight-bladed β-propeller domain (amino acids 55–497) consists of four-stranded antiparallel β-sheet motifs containing a short helix with a double glutamate residues motif (Glu205 and 206), inserted into the first sheet of the fourth propeller blade, and an S2-loop (Ser349-Glu361). The α/β-hydrolase domain contains a central β-sheet constituted by eight β-strands (six parallel and two antiparallels) surrounded by 12 α-helices, assembled by residues 506–766 at the C-terminus, and a short stretch of the N-terminal sequence (residues 39–51).11,12 The C-terminal region contains a hydrophobic pocket (Tyr547, Ser630, Tyr631, Val656, Trp659, Asp663, Tyr666, Asn710, Val711 and Gly740). The active site, located in a large cavity at the interface of the two domains, contains a catalytic triad formed by Ser630, Asp708, and His740. The C-terminal loop of DPP-IV is essential for both dimer formation and optimal catalytic efficacy. Five disulfide bridges stabilize the protein: four are present in the β-propeller region between Cys444/447, 385/394, 454/472, and 328/339, and the last at the α/β-hydrolase domain between Cys649 and 762.11


image file: d3np00007a-f1.tif
Fig. 1 Ribbon diagram showing the overall structure of the DPP-IV homodimer, viewed perpendicular to the twofold dyad axis, complexed with 4-(2,4,5-trifluorophenyl)butane-1,3-diamine inhibitor (https://www.rcsb.org/structure/4j3j).

DPP-IV is produced as a monomer; nevertheless, it needs dimerization for its proteolytic activity. The body has two DPP-IV isoforms: the membrane-bound DPP-IV, composed of full-length amino acids, and soluble DPP-IV (residues 49–766), whose cytoplasmic and transmembrane regions are absent. The soluble form exerts its proteolytic action by clipping dipeptides from the penultimate position of its substrates.

Most inhibitors bind to the DPP-IV-GLP-1 interacting site as protein–protein interaction inhibitors. Residues from the hydrolase and propeller domains participate in binding some inhibitors, such as valine–pyrrolidine and a few gliptins. The Glu residues 205 and 206 form salt bridges to the free amino group of the substrates. Glu205 and Glu206 are situated in a small horizontal helix (residues 201–207), which narrows the active site, leaving room for only two amino acids, making DPP-IV a dipeptidyl peptidase. The spatial arrangement of serine 630 (active site) to glutamic acids 205 and 206 in DPP-IV makes these two amino acid residues the most critical feature for the alignment of the peptide before cleavage. According to the site of binding, the commercial DPP-IV inhibitors belong to three classes (Fig. 2).13


image file: d3np00007a-f2.tif
Fig. 2 Marketed drugs with DPP-IV inhibitory activities.

Class 1 (e.g., vildagliptin and saxagliptin) interacts with subsites S1 (a hydrophobic pocket containing Ser630, Val656, Trp659, Tyr662, Val711, and Asn710) and S2 (Arg125, Arg669, Glu205 and 206, Phe357, and Arg358). In the case of saxagliptin, a few studies, including NMR, suggest that its inhibition of DPP-IV most likely involves a reversible covalent enzyme–inhibitor complex formation.

Class 2 (e.g., alogliptin and linagliptin) attach to subsites S2, S1, S1′ (Phe357, Tyr547, Pro550, Ser630, and Tyr residues 631 and 666), and S2′ (Tyr547, Trp629, Ser630 and His740).

Class 3 (e.g., sitagliptin and teneligliptin) binds to extensive site S2 (Phe357, Arg358, Ser209, and Val207), S2, and S1 subsites. In general, the N-terminus of the substrate peptide or inhibitor is recognized by Glu205, Glu206, and Ser630 cleaves at the N-terminus penultimate position. Glu205 and Glu206 amino acids form salt bridges and hydrogen bonds with the ligands.

For class 2 and 3 inhibitors, interactions with Tyr547 and Trp629 and with Phe357 and Tyr666, respectively, are pivotal for their DPP-IV inhibitory activities. X-ray analyses have determined the structures of DPP-IV and DPP-IV-inhibitors complexes.14

3.2. Implication of DPP-IV in diabetes

The L-cells of the gut release the incretin hormone GLP-1 within minutes in response to food intake in two primary biological forms: GLP-1 amide and GLP-1. The latter is the most abundant under physiological conditions. Soluble DPP-IV rapidly degrades GLP-1 (half-life shorter than 2 min) into inactive GLP-1 amide. GLP-1 induced an increment of intracellular cAMP upon binding to its G-protein-coupled receptor on the pancreatic β-cells (Fig. 3). Then, after downstream signaling is accomplished, insulin secretion increases (incretin effect) and regulates postprandial glucose. In addition, GLP-1 inhibits glucagon secretion, regulates satiety and appetite, and increases α- and β-cell insulin sensitivity and gene expression. In diabetic patients, DPP-IV is widely expressed at the surface of epithelial and endothelial cells, causing the rapid degradation of GLP-1 (7–36) to GLP-1 (9–36) (Fig. 3).15–19
image file: d3np00007a-f3.tif
Fig. 3 Intestine and pancreatic endocrine pathways altered during DPP-IV inhibition.

On the other hand, GIP is synthesized and secreted from K-cells of intestinal epithelium located in the proximal duodenum. DPP-IV terminates its insulinotropic action, cleaving the N-terminal dipeptide of GIP (1–42) to inactive GIP (3–42). GIP in adipose tissue stimulates fatty acid synthesis, enhances insulin-stimulated incorporation of fatty acids into triglycerides, increases insulin receptor affinity, and increases insulin-stimulated glucose transport. Hence, GIP is considered more relevant for treating obesity and does not affect glucagon secretion (Fig. 3).19,20

Concerning TIIDM, GLP-1 is more relevant because it accelerates glucose-induced insulin secretion. The increment of the half-lives of GLP-1 and GIP by DPP-IV inhibitors lowers fasting and postprandial hyperglycemia due to the increment of insulin secretion in a glucose-dependent manner and a decrease of excessive glucagon secretion observed in TIIDM. In addition, DPP-IV inhibitors are weight-neutral and well-tolerated.18,19

3.3. Assay methods

In general, drug screening for DPP-IV inhibitors has followed a target-based strategy using enzymatic assays, which give a precise mode of action. Unfortunately, these assays cannot detect off-target or undesirable pharmacological effects. The assays to discover DPP-IV inhibitors use recombinant enzymes, usually human or porcine, combined with colorimetric or fluorometric detection methods.21,22 The procedures are rapid, simple, sensitive, reliable, and usually performed with commercial kits. The colorimetric assay is based on the hydrolysis by DPP-IV of Gly-Pro-p-nitroanilide to yield a constant chromophore (4-nitroaniline) in the presence of the potential inhibitor.21 The production of 4-nitroaniline is quantified using UV-VIS spectroscopy in a microplate reader at λmax 360 and 460 nm. The fluorometric assay uses a fluorogenic substrate [Gly-Pro-aminomethylcoumarin (AMC)] to measure DPP-IV activity. Cleavage of the peptide bond by DPP-IV releases the AMC group, resulting in fluorescence that can be measured using an excitation wavelength of 350–360 nm and an emission wavelength of 450–465 nm. In addition, other fluorescent probes with the function of aggregation-induced emission have been used to screen DPP-IV inhibitors.23 The results for these bioassays are usually expressed as % of inhibition or IC50 values.

Simple and rapid colorimetric and electrochemical methods using gold nanoparticles as the probe have been developed to discover DPP-IV inhibitors; however, they lack sensitivity.24,25 Other methods for detecting DPP-IV inhibitors are liquid chromatography coupled to mass spectrometry and thin-layer chromatography-bioautography-mass spectrometry combined with a liquid chromatography-mass spectrometry-controlled purification system.26–28

Over the past few years, in silico techniques (quantitative structure–activity relationship, QSAR), ligand-based and structure-based virtual screening, pharmacophore modeling, molecular docking, molecular dynamic, and ADMET predictions have been widely used in the search for compounds with DPP-IV inhibitory properties.29,30

Finally, bioassays based on cell cultures or small animal studies, which are more relevant to humans, are currently being developed. In most assays, diprotin A (IPI), hemifumarate, or some gliptins are employed as positive controls.

3.4. Natural products with DPP-IV inhibitory properties

Natural DPP-IV inhibitors have been the topic of several review articles.31–40 Some of these reviews describe mainly the testing of plant extracts or in silico studies.29,30,37,40,41 The evaluation of plant extracts is beneficial from the point of view of traditional medical practices, in which the efficacy of the treatments is due to the synergistic action of the mixture of compounds present in an extract (polypharmacy) or the combination of several herbs. An excellent review regarding the inhibitory action on DPP-IV activity of plant-based polyherbal formulations and nano phytomedicines was recently published.37 Therefore, a comprehensive review focusing solely on natural enzymatic inhibitors would benefit the community. The natural DPP-IV inhibitors discovered during 2000–2022 are summarized and categorized in Tables 1–4 These include the compound type and name, inhibitory effect, positive control used in the enzymatic evaluation, natural sources, and the corresponding references. Unfortunately, the diversity of non-standardized assays employed, the lack of positive control in some studies, and the non-disclosure of inhibitors' concentration range used in evaluations preclude any critical structure–activity relationship analysis.
3.4.1. Flavonoids and other aromatic compounds. Flavonoids are the most abundant plant polyphenols consumed regularly with regular food intake or in natural extracts used medicinally. These polyphenols regulate DPP-IV activity and thus exert their antidiabetic effects. Representative DPP-IV inhibitory flavonoids (1–72) and other aromatic compounds (73–100) are summarized in Table 1.42–71
image file: d3np00007a-u1.tif

image file: d3np00007a-u2.tif

image file: d3np00007a-u3.tif

image file: d3np00007a-u4.tif

image file: d3np00007a-u5.tif

image file: d3np00007a-u6.tif

image file: d3np00007a-u7.tif

image file: d3np00007a-u8.tif
Table 1 Representative flavonoids and other aromatic compounds with DPP-IV inhibitory activitya
Compound IC50 (μM) or inhibition (%) Control, IC50 (μM) or inhibition (%) Source Ref.
a IPI: diprotin A; SG: sitagliptin; ALG: alogliptin; RA: rosmarinic acid; LG: linagliptin; HF: hemifumarate; NM; not mentioned; CT: commercial standard.
Anthocyanins
Cyanidin 3-O-β-D-glucopyranoside (1) 0.4 IPI, 4.2 Berries 42
8.3 SG, 0.1 CT 43
138.8 NM CT 44
Cyanidin (2) 1.4 IPI, 4.2 Berries 42
Malvidin (3) 1.4 IPI, 4.2 Berries 42
[thin space (1/6-em)]
Aurones
Aureusidin 6-O-β-D-glucopyranoside (4) 24.3 ALG, 0.002; IPI, 2.3 Helichrysum arenarium (L.) Moench (Asteraceae) 45
Leptosidin (5) 13.3 SG, 0.1 Coreopsis lanceolata L. (Asteraceae) 46
[thin space (1/6-em)]
Catechins
Catechin (6) 175.0 SG, 0.1 CT 43
(−)-Epicatechin (7) 290[thin space (1/6-em)]260.0 SG, 60[thin space (1/6-em)]174.7 Tetracera indica Merr. (Dilleniaceae) 47
Epigallocatechin-3-O-(3-O-methyl) gallate (8) 75% at 100 μM SG, 80% at 50 μM Camellia sinensis L. (Theaceae) 48
206.0 SG, 0.1 CT 43
[thin space (1/6-em)]
Chalcones
3,2′-Dihydroxy-4-3′-dimethoxychalcone-4′-O-β-D-glucopyranoside (9) 14.3 SG, 0.1 C. lanceolata L. (Asteraceae) 46
Arenariumoside III (10) 54.1 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
Chalconaringenin 2′-O-β-D-glucopyranoside (11) 23.1
Chalconaringenin 2′,4′-di-O-β-D-glucopyranoside (12) 70.0
Lanceoletin (13) 9.6 SG, 0.1 C. lanceolata L. (Asteraceae) 46
4-Methoxylanceoletin (14) 21.6
Phloretin (15) 139.0 SG, 0.1 CT
CT 43
Isoliquiritigenin (16) 150.0 NM 44
[thin space (1/6-em)]
Flavanones
(2R)-8-Methoxybutin (17) 64.9 SG, 0.1 C. lanceolata L. (Asteraceae) 46
Eriocitrin (18) 10.4 IPI, 4.2 Citrus spp (Rutaceae) 42
Eriodictyol (19) >20.0 SG, 0.1; IPI, 31.0 CT 49
0.5 SG, 0.1 Lippia graveolens Kunth (Verberaceae) 50
10.9 RA, 14.1 CT 51
142.0 SG, 0.1 CT 43
Hesperidin (20) 0.3 IPI, 4.2 Citrus spp (Rutaceae) 42
157.0 SG, 0.1 CT 43
Naringenin (21) >20.0 SG, 0.1; IPI, 31.0 CT 49
10.9 SG, 0.1 L. graveolens Kunth (Verberaceae) 50
2.5 RA, 14.1 Salvia spp. (Lamiaceae) 51
0.2 IPI, 4.2 Citrus spp (Rutaceae) 42
[thin space (1/6-em)]
Flavanonols
Dihydromyricetin (22) 107.0 SG, 0.1 CT 43
Taxifolin (23) >20.0 SG, 0.1 CT 50 and 51
6.7 IPI, 7.2 CT 52
188.0 SG, 0.1 CT 43
[thin space (1/6-em)]
Flavones
Cirsimaritin (24) 0.4 SG, 0.1 CT 51
2.5 SG, 0.1 L. graveolens Kunth (Verberaceae) 50
Chrysin (25) 33% at 200 μM SG, 0.1; IPI, 31.0 CT 49
150.0 SG, 0.1 CT 43
Chrysoeriol (26) 37% at 200 μM SG, 0.1; IPI, 31.0 CT 49
12.4 SG, 0.1 CT 43
Baicalein (27) 43% at 200 μM SG, 0.1; IPI, 31.0 CT 49
48.5 SG, 0.1 CT 43
Acacetin (28) >20.0 SG, 0.1; IPI, 31.0 CT 49
Apigenin (29) 442% at 200 μM
0.1 IPI, 4.2 Citrus spp (Rutaceae) 42
28.1 SG, 0.1 CT 43
Apigenin 4′-O-β-D-glucopyranoside (30) 182[thin space (1/6-em)]500.0 SG, 1800.0 Paeonia delavayi Franchet (Paeoniaceae) 53
62.7 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
Apigenin 7-O-β-D-glucopyranoside (31) 80.0 SG, 0.1 CT 43
Apigenin 7-O-β-D-glucopyranosiduronic acid methyl ester (32) 50.5 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
Apigenin 7-O-β-D-gentiobioside (33) 32.0 ALG, 0.002; IPI, 2.3 45
Flavone (34) 0.2 IPI, 4.2 Citrus spp (Rutaceae) 42
Genistein (35) 0.5 Glycine max (L.) Merr (Fabaceae) 42
162.0 SG, 0.1 CT 43
Hispidulin (36) 46% at 200 μM SG, 0.1; IPI, 31.0 CT 49
0.5 SG, 0.1 CT 50 and 51
0.5 SG, 0.7 Lens culinaris Medikus (Fabaceae) 46 and 54
6-Hydroxyluteolin 7-O-β-D-glucopyranoside (37) 60.3 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
6-Hydroxy-3′-O-methylluteolin-7-O-β-D-glucopyranoside (38) 73.2
Luteolin (39) 45% at 200 μM SG, 0.1; IPI, 31.0 CT 49
0.1 IPI, 4.2 Citrus spp (Rutaceae) 42
15.0 SG, 0.1 CT 43
Luteolin 7-O-β-D-glucopyranoside (40) 37.5 ALG, 0.002; IPI, 2.3 CT 45
39.6 SG, 0.1 CT 43
Breviscapin (41) 49.5 SG, 0.2 Scutellaria baicalensis Georgi (Lamiaceae) 55
Isoschaftoside (42) 108.0 SG, 0.1 CT 43
Vitexin (43) 60.3
33.1 LG, Smilax china L. (Smilacaceae) 56
Vitexin 4′-O-β-D-glucopyranoside (44) 65.2 SG, 0.1 CT 43
[thin space (1/6-em)]
Isoflavone
Formononetin (45) 99.1 SG, 0.1 CT 43
[thin space (1/6-em)]
Flavonols
Fisetin (46) 54.2 SG, 0.1 CT 43
Galangin (47) 35% at 200 μM SG, 0.1; IPI, 31.0 CT 49
40.1 NM CT 57
207.0 SG, 0.1 CT 43
Guaijaverin (48) 86.0 HF, 0.1 CT 58
Lepidoside (49) 43.6 LG, 0.1 S. china L. (Smilacaceae) 56
Morin (50) 37% at 200 μM SG, 0.1; IPI, 31.0 CT 49
Rutin (51) 40% at 200 μM SG, 0.1; IPI, 31.0 CT 49
>100.0 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
32.9 LG, 0.1 S. china L. (Smilacaceae) 56
312.0 SG, 0.1 CT 43
Robinin (52) 37.0 SG, 0.7 L. culinaris Medikus (Fabaceae) 46 and 54
Isorhamnetin (53) 60.3 SG, 0.1 CT 43
Isorhamnetin 3-O-β-D-glucopyranoside (54) 23.7 SG, 0.3 Capsicum frutescens L. (Solanaceae) 59
6.5 SG, 0.1 CT 43
Narcissoside (55) 166.5 NM CT 44
8.6 SG, 0.1 CT 43
Kaempferol (56) 38% at 200 μM SG, 0.1; IPI, 31.0 CT 49
0.5 IPI, 4.2 Citrus spp (Rutaceae) 42
51.9 SG, 0.7 L. culinaris Medikus (Fabaceae) 46 and 54
46.0 LG, 0.1 S. china L. (Smilacaceae) 56
165.0 SG, 0.1 CT 43
160[thin space (1/6-em)]465.4 SG, 60[thin space (1/6-em)]174.7 T. indica Merr. (Dilleniaceae) 47
Kaempferol 3-O-rutinoside (57) 65% at 100 μM SG, 80% at 50 μM C. sinensis L. (Theaceae) 48
Kaempferol 3-O-β-gulcopyranosyl-(1→2)-β-galactopyranosyl-7-O-α-rhamnopyranoside (58) 27.9 SG, 0.7 L. culinaris Medikus (Fabaceae) 46 and 54
Kaempferol 3-O-β-glucopyranosyl-(1→2)-[α-rhamnopyranosyl-(1→6)]-β-galactopyranosyl-7-O-α-rhamnopyranoside (59) 36.5 SG, 0.7 L. culinaris Medikus (Fabaceae) 46 and 54
Kaempferol 3-O-β-glucopyranosyl -(1→ 6)-glucopiranoside (60) 58.0 ALG, 0.002 IPI, 2.3 CT 45
Kaempferol 3,7-di-O-β-D-glucopyranoside (61) 36.8 ALG, 0.002 IPI, 2.3 CT 45
Kaempferol 3,4′-di-O-β-D-glucopyranoside (62) 88.6
Kaempferol 3-O-β-D-glucopyranosyl-(1→ 3)-O-β-D-glucopyranoside (63) 85.2
Kaempferol 7-O-α-L-rhamnoside (64) 20.8 LG, 0.1 S. china L. (Smilacaceae) 56
Myricetin (65) 15.1 SG, 0.1 CT 43
156.3 NM CT 44
Myricetin 3-O-glucoside/galactoside (66) 90% at 100 μM SG, 80% at 50 μM C. sinensis L. (Theaceae) 48
Myricitrin (67) 47.2 SG, 0.1 CT 43
Quercetin (68) 46% at 200 μM SG, 0.1; IPI, 31.0 CT 49
2.9 IPI, 4.2 Citrus spp (Rutaceae) 42
8.3 IPI, 3.2 CT 60
4.0 SG, 5.5; IPI, 0.7 CT 61
145.0 SG, 0.1 CT 43
147[thin space (1/6-em)]300.0 SG, 1800.0 P. delavayi Franchet (Paeoniaceae) 53
71[thin space (1/6-em)]972.2 SG, 60[thin space (1/6-em)]174.7 T. indica Merr. (Dilleniaceae) 47
Quercetin 3-O-β-D-glucopyranoside (69) 56.6 ALG, 0.002 IPI, 2.3 CT 45
96.8 NM CT 44
Hyperoside (70) 138.8 NM CT 44
Quercetin 3,3′-di-O-β-D-glucopyranoside (71) 73.9 ALG, 0.002; IPI, 2.3 CT 45
Quercitrin (72) 160.1 SG, 0.3 Rhus chinensis Mill. (Anacardiaceae) 62
[thin space (1/6-em)]
Other aromatic compounds
3,6′-O-Diferuloylsucrose (73) 46.2 SG, 0.07 Lilium longiflorum Thunb. (Liliaceae) 63
4-O-Acetyl-3,6′-O-diferuloylsucrose (74) 63.3
Arenariumoside V (75) 83.0 ALG, 0.002; IPI, 2.3 H. arenarium (L.) Moench (Asteraceae) 45
Arenariumoside VI (76) 52.0
Arenariumoside VII (77) 83.0
Caffeic acid (78) 3.4 IPI, 4.2 CT 42
Calebin A (79) 6.1 SG, 83% at 100 μM CT 64
Chlorogenic acid (80) 124.5 SG, 0.1 CT 65
233.7 SG, 0.3 CT 66
0.8 IPI, 1.1 CT 67
Coumarin (81) 54.8 SG, 5.5; IPI, 0.7 CT 61
Emodin (82) 5.8 SG, 21.8 Rheum palmatum Baill. (Polygonaceae) 68
Gallic acid (83) 4.7 IPI, 4.2 CT 42
Mangiferin (84) 44.2 SG, 0.2 Mangifera indica L. (Anacardiaceae) 55
Methyl p-coumarate (85) 911.4 SG, 34.0 Melicope latifolia T.G.Hartley (Rutaceae) 69
Resveratrol (86) 0.001 IPI, 4.2 Vitis vinifera L. (Vitaceae) 42
27.3 SG, 0.1 V. vinifera L. (Vitaceae) 70
5.6 IPI, 7.2 CT 52
14.1 IPI, 3.3 CT 60
611[thin space (1/6-em)]800.0 IPI, 11[thin space (1/6-em)]100.0 Senna siamea Lam. (Fabaceae) 71
Piceatannol (87) 22.0 SG, 0.1 V. vinifera L. (Vitaceae) 70
Ovyresveratrol (88) 28.7 V. vinifera L. (Vitaceae)
2-Prenyl reverastrol (89) 22.1 Propolis
4-Prenyl resveratrol (90) 21.3 Arachis hypogaea L. (Fabaceae)
2-Prenyl piceatannol (91) 21.9 Propolis
2-Prenyl oxyresveratrol (92) 23.8 Cudrania tricuspidate Carriere (Moraceae)
4-Prenyl oxyresveratrol (93) 24.1 Artocarpus integer Spreng (Moraceae)
2-Geranyl resveratrol (94) 17.7 Macaranga trichocarpa (Zoll.) Müll.Arg. (Euphorbiaceae)
4-Geranyl resveratrol (95) 22.8 Chlorophora excelsa Welw (Moraceae)
2-Geranyl piceatannol (96) 4.6 M. trichocarpa (Zoll.) Müll.Arg. (Euphorbiaceae)
4-Geranyl piceatannol (pawhuskin C) (97) 3.9 Dalea purpurea Vent. (Fabaceae)
4-Geranyl oxyresveratrol (chlorophorin) (98) 9.1 Artocarpus xanthocarpus Forst (Moraceae)
Rosmarinic acid (99) 0.4 SG, 0.1 Rosmarinus officinalis Schleid. (Lamiaceae) 50
Theogallin (100) 20% at 100 μM SG, 80% at 50 nM C. sinensis L. (Theaceae) 48


Perhaps, one of the most extensive analyses of the effect of flavonoids on DPP-IV was carried out by Pan et al.,44 who analyzed 70 different flavonoids of different structural groups using a human recombinant enzyme and monitoring the reaction by measuring the fluorescence generated by the substrate Gly-Pro-7-amido-4-methyl coumarin hydrobromide.

The most relevant aspects of this study are briefly discussed in the next paragraph. Cyanidin 3-O-β-D-glucopyranoside (1), isoliquiritigenin (16), narcissoside (55), myricetin (65), and hyperoside (70) inhibited the enzyme higher than 50% at 200 μM. The IC50 values of these flavonoids were 81.05, 149.96, 166.52, 156.29, and 138.79 μM, respectively. The authors concluded that the inhibitory effect on DPP-IV is better when the flavonoids are hydroxylated at C-6 of ring A and ring B. Methylation of these phenolic groups reduces the activity. Hydroxylation at C-3 diminishes the activity, but its methylation improves the inhibitory action. The 2,3-double bond in conjugation with the 4-carbonyl group in ring C is relevant for inhibiting DPP-IV. Thus, flavones and flavanols are more active than the corresponding flavanones and flavanonols. The influence of glycosylation depends on the flavonoid skeleton and the point of attachment of the sugar unit. Hence, apigenin (29), luteolin (39), and myricetin (65) show better inhibitory effects than their glycosides, while the inhibitory activities of isorhamnetin (53) and kaempferol (56) and were smaller than the corresponding glycosides. Proença et al.39 reported similar results; they based their conclusion on evaluating 140 flavonoids (natural and synthetic) using in vitro and ex vivo models. They also demonstrated that the fluorometric method is more sensitive than the colorimetric.39,72

Regrettably, unlike Proença et al.,39 Pan et al.,44 did not include the results for any positive control in the study; therefore, potency comparisons between such control and the tested flavonoids cannot be performed. In the study conducted by the former authors, natural flavonoids did not reach 50% inhibition at 200 μM and were thus less effective and potent than positive controls assayed (sitagliptin and diprotin A with IC50 values of 0.064 and 31 μM, respectively).

Kinetic analyses of cyanidin 3-O-β-D-glucopyranoside (1), isoliquiritigenin (16), narcissoside (55), and hyperoside (70) revealed that they were mixed-type inhibitors; furthermore, their competitive inhibition constants were lower than the uncompetitive ones. Thus, like most flavonoids, these compounds preferred interacting with the enzyme's active site. In addition, their docking studies predicted their binding with similar amino acids as vildagliptin and sitagliptin, behaving, thus, as class 1 inhibitors. Myricetin (65) was the only compound that acted as a noncompetitive inhibitor. Other authors reported that apigenin (29), the synthetic 2-(3,4-dimethoxyphenyl)-3,7,8-trihydroxy-4H-chromen-4-one72 and luteolin (39)42 are noncompetitive inhibitors of DPP-IV. Flavone (34), although less active, was reported by Fan et al.42 as a competitive inhibitor, and the docking analysis of the latter compound supported its behavior as a class 2 inhibitor. Pan et al.44 also supported their finding with docking studies since myricetin (65) interacts with different residues than 1, 16, 55, and 70. The interactions of 65 were also different from those of inhibitors belonging to class 1–3. The relevant difference between myricetin (65) and 1, 16, 55, and 70, other than the basic scaffold, was the higher level of hydroxylation in ring B.

On the other hand, Gao et al.43 compared the in vitro inhibitory activity of 30 dietary flavonoids on DPP-IV. They found that isorhamnetin-3-O-β-D-glucoside (54, IC50, 6.53 μM), cyanidin-3-O-β-D-glucoside (2, IC50, 8.26 μM), and narcisoside (55, IC50, 8.57 μM) were the most active; sitagliptin was used as positive control (mean IC50, 0.12 μM). Moreover, the three flavonoids suppress DPP-IV activity and expression in Caco-2 cells similarly to sitagliptin. These authors conducted a QSAR analysis and found that minor and electron-withdrawing groups at positions 4′ of ring B and 5 and 7 of ring A could improve DPP-IV inhibition. They also reported docking studies of 1 and 55, which differed entirely from Pan et al.44 results. Unfortunately, Gao et al.43 did not conduct a kinetic analysis, which precludes further comparison.

Some of the DPP-IV flavonoid inhibitors affect glucose homeostasis through different mechanisms according to the human, animal, and in vitro studies.7,24,31,37,73 For example, anthocyanins, in general, modify the activities of glucose transporter-4 (GLUT-4), SGLTs, α-glucosidase, protein tyrosine phosphatase 1B (PTP-1B), GLP-1/GLP-1R and peroxisome proliferator-activated receptor-γ.44,74 Catechins decrease blood glucose levels by activating glucose transporters and the insulin receptor. Some flavanones and flavones, such as hesperidin (20) and genistein (35) control blood glucose levels by acting through different mechanisms. Both compounds modify the activity of gluconeogenic enzymes and activate the lipogenic processes. Rutin (51), myricetin (65) and quercetin (68) are hypoglycemic compounds in vivo, and increase glucose uptake and tolerance.73,74

Other polyphenols have been evaluated using enzymatic assays (Table 1), and only in a few cases have structure–activity relationships or kinetic studies been conducted.

Emodin (82), an anthraquinone, rosmarinic acid (99), a phenylpropanoid, and some stilbenoids are among the most active polyphenols; some of these compounds have shown antidiabetic effects in vivo and in vitro targeting other enzymes and receptors involved in glucose metabolism.

Resveratrol (86) was a competitive inhibitor of DPP-IV, and the docking analysis predicted that it could be a Class 2 inhibitor. According to a study by Bo et al.,70 resveratrol (86), piceatannol (87), oxyreverastrol (88), and their prenylated analogs showed similar IC50 values ranging from 11.47 to 28.67 μM excepting the geranyl derivatives, which exhibited the highest DPP-IV inhibitory activity. These results indicate that geranyl substitution at the B ring and hydroxylation at the 3′ positions of resveratrol are critical in DPP-IV inhibitory activity. Although no kinetic analysis was performed in this study, docking investigation of pawhuskin A (97, 4-geranyl piceatannol) with DPP-IV revealed that the core skeleton of piceatannol interacted with various amino acid residues, including Glu703, Lys87, and Asp704. In contrast, the geranyl group interacted with Trp89, Ser92, Tyr176, Val217, Phe205, and Asp674. Moreover, it was observed that three hydrogen bonds were formed between the hydroxyl groups of 4-geranyl piceatannol (97) and three DPP-IV (Gln88, Ser709, and Ala708) amino acid residues. Resveratrol (86) targets other enzymes and physiological processes involved in TIIDM, as reviewed by a few authors.66,74,75

Emodin (82) is the most studied anthraquinone as a potential therapeutic agent against TIIDM using cell cultures and animal models. The anthraquinone 82 activates 5′-adenosine monophosphate-AMPK, boosts glucose uptake, regulates glucose utilization in several tissues and ameliorates high-fat-diet-induced insulin resistance by reducing lipid accumulation through decreasing fatty acid transport protein 1 in rat skeletal muscle. Moreover, emodin impacts inflammation processes inhibiting the release of tumor necrosis factor α.76

Chlorogenic acid (80), a polyphenol widely distributed in plants, is the only uncompetitive inhibitor reported in this group of compounds.66,67 It also normalized the metabolism of glucose and lipids by increasing the expression of GLUT-4 by different mechanisms including inhibition of G6Pase and suppressing glucose transport in the intestine. Gallic acid (83) also reduced triglyceride and total cholesterol.74

Finally, rosmarinic acid (99) reduces intestinal glucose absorption, inhibits PTP-1B, activates AMPK, and stimulates glucose uptake by translocation of GLUT-2 and GLUT-4; mainly, these mechanisms are consistent with the hypoglycemic effects of compound 99in vivo.77

3.4.2. Peptides. As flavonoids, peptides have been the subject of numerous investigations. There are many animals (tuna, eggs, dairy products, salmon skin, among others) and vegetables (soybean, rice, amaranth, draft beer, broccoli, Brassica spp, macroalgae, yam species, and beans) proteins, which yield inhibitory DPP-IV peptides. In this review, we will focus only on those peptides originating from vegetal dietary proteins, which according to a few investigations, can inhibit DPP-IV in vitro.78–81 The most active peptides found in this review are summarized in Table 2,82–91 which includes amino acid sequences, inhibitory activities, positive controls, and natural sources.
Table 2 Representative peptides isolated from vegetal species with DPP-IV inhibitory activitya
Source Sequence IC50 (μM) Control, IC50 (μM) or inhibition (%) Ref.
a IPI: diprotin A; SG: sitagliptin; NM; not mentioned. *Values expressed in mg mL−1.
Yam dioscorin [Dioscorea spp (Dioscoreacea)] RRDY (101) 930.0 SG 34% at 0.03 μM 81 and 90
IHF (102) 3770.0
KRIHF (103) 4110.0
RL (104) 1200.0
GPA (105) 2870.0
MGSF (106) 2120.0
DPF (107) 1540.0
Common bean [Phaseolus vulgaris L. (Fabaceae)] KTYGL (108) 0.03* IPI, 58.6 87
KKSSG (109) 0.6*
CPGNK (110) 0.9*
GGGLHK (111) 0.6*
Draft beer LNFDPNR (112) 471.5 NM 82
LPQQQAQFK (113) 489.9
Broccoli [Brassica oleracea Plenck (Brassicaceae)] LPGVLPVA (114) 392.0 NM 83
YLYSPAY (115) 181.0
Brassica napus L. (Brassicaceae) ELHQEEPL (116) 78.5 IPI, 3.6 84
EL (117) 185.0
HQEEP (118) 97.3
Rice bran [Oryza sativa L. (Poaceae)] IP (119) 410.0 IPI, 210.0 82 and 83
MP (120) 870.0
VP (121) 880.0
RP (122) 224.0
TP (123) 237.0
LP (124) 237.0
KP (125) 254.0
HP (126) 282.0
YP (127) 317.0
FP (128) 363.0
WP (129) 453.0
PP (130) 586.0
SP (131) 598.0
AP (132) 795.0
LAHKALCSEKL (133) 165.0
TKCEVFRE (134) 166.0
LCSEKLDQ (135) 186.0
Palmaria palmata (L.) Kuntze (Palmariaceae) ILAP (136) 43.4 IPI, 3.6 88
LLAP (137) 53.7
MAGVDHI (138) 159.4
Soybean glycinin [Glycine max (L.) Merr (Fabaceae)] IAVPTGVA (139) 106.0 SG, 88% at 0.1 μM 81 and 89
KL (140) 159.8
LR (141) 2083.6
Lupin seed [Lupinus albus L. (Fabaceae)] LTFPGSAED (142) 228.0
Wheat gluten hydrolysates [Triticum aestivum L. (Poaceae)] VPL (143) 15.8 IPI, 4.0 91
WP (144) 45.0
QPG (145) 70.9 NM
QPF (146) 71.7
SPQ (147) 78.9
QPQ (148) 79.8
LPQ (149) 56.7
IA (150) 88.0


After optimized enzymatic hydrolysis with different gastrointestinal proteases, in combination with bioassay-guided fractionation, food vegetal proteins are degraded to yield DPP-IV inhibitory peptides. The procedures for hydrolysis, fractionation, purification, and identification of these DPP-IV inhibitory peptides have been the subject of a recent review.81 The identification of the peptides is based on tandem mass (MS/MS) or Edman degradation of the purified peptides. Afterward, the peptides characterized from the active fractions are synthesized and tested in vitro.

Recently, a novel strategy combining liquid chromatography-tandem mass spectrometry (LC-MS/MS) and in silico analysis has become more efficient. This procedure involves three steps: (i) Nano-LC-MS/MS for peptide identification in complex mixtures like protein hydrolysates; (ii) molecular docking or quantitative structure–activity relationship (QSAR) models based in silico analysis for indicating the relationship between identified or known peptides and target proteins, which may be helpful to screen some active peptides from those identified; and (iii) the potentially active peptides obtained based on virtual screening should be synthesized, and their efficacy determined in vitro or in vivo.

Bioinformatics is used in protein and peptide research and has become a powerful tool that can be used for in silico prediction of potentially bioactive peptides released from food proteins. For example, the BIOPEP database is a bioinformatics tool enabling the detection of biologically active fragments in protein sequences and the classification of proteins as potential sources of bioactive molecules. The simulation of protein hydrolysis to find peptides that can be released by a given enzyme or as a result of the combined action of two, three, or more enzymes. For more details, see the work of Liu et al.81

According to Nongonierma and Fitzgerald,91 active dipeptides have W, T, or M at their N-terminus; and A, L, or H at their C-terminus. In the case of tripeptides, the amino acid more frequently found in the N-terminus are I, Q, L, S, or V. In contrast, in the C-terminus, the last amino acid could be Q, A, I, L, G, M or F, and in all cases the middle amino acid is P. Finally, longer peptides frequently possess L/G/I, P/L/K, A/V/G/P at positions 1, 2, and 3, and P/L/R at their C-terminus.

IPI and diprotin B (VPL) are among the most important DPP-IV inhibitors (IC50 1.1 and 5.5 μg mL−1, respectively). Both tripeptides were first isolated in 1984 from culture filtrates of Bacillus cereus BMF673-RF1.92 The structure of the human DPP-IV-IPI complex was analyzed by X-ray crystallography; in the complex the conformation of both monomers of the enzyme is nearly the same as without IPI. This tripeptide binds at the S2, S1, and S1′ sites, through hydrogen bond (Glu205, Glu206, Tyr662, Tyr547, Tyr631, Arg125), electrostatic (Arg125) and hydrophobic (Tyr666, Phe357, Val656, Tyr662, Tyr63, Trp659, Tyr547) interactions.93 Rahfeld et al.,94 however, concluded that it is better to refer to IPI, VPL, and other tripeptides with an analogous structure as substrates for DPP-IV. They found that the apparent competitive inhibition by these compounds is a kinetic artifact, which comes from the substrate-like nature of tripeptides with a penultimate proline residue.91

IPI has also been isolated from Gynura divaricata (L.) DC (Asteraceae), a Chinese herbal medicine widely grown and used to treat diabetes in China and Thailand. The hypoglycemic activity of G. divaricata has been demonstrated in various reports. Several diprotin analogs have been characterized in the same plant using UHPLC-HRMS combined with a library search. The fragmentation patterns associated with molecular networking allowed complete identification, and docking analysis of these peptides revealed their potential DPP-IV inhibitory activity.95 The IPI fragment is also present in the primary sequence of several peptides obtained from the hydrolysis of food proteins (amaranth, soybean, wheat, maize, and rice).91 Amaranth is relevant for Latin American countries, where it has been a traditional food in some pre-Columbian civilizations. Its high nutritional value is due to its high lysine and methionine content.96 Velarde et al.96 validated that the mixtures of peptides from amaranth tryptic glutelins hydrolysates inhibited DPP-IV, with an IC50 value of 1.1 mg mL−1. Unfortunately, the single peptides have not been tested in vitro, but by docking modeling.

Pepsin hydrolysis of yam dioscorin yielded a few inhibitory peptides against DPP-IV. The most active peptides, namely RRDY (101), RL (104), and DPF (107), were further investigated in normal ICR mice using an oral glucose tolerance test (OGTT). The peptides, sitagliptin (positive control), or yam protein were administered orally. The result revealed that only RRDY (101, 100 mg kg−1), yam dioscorin (100 mg kg−1), or sitagliptin (10 mg kg−1) decreased the postprandial glucose peak upon the carbohydrate charge and increased the level of blood insulin, consistent with an inhibitory effect of the DPP-IV, without ruling out that other mechanisms could be involved in the observed pharmacological effect.90

Mojica et al.97 reported kinetic studies of peptides KTYGL (108), KKSSG (109), CPGNK (110), and GGGLHK (111) obtained from common bean protein digests; the four peptides were competitive inhibitors of DPP-IV, which agreed with the docking studies performed by the same authors, which predicted that the peptides could interact with several residues in different sites of the enzyme blocking the access of the substrate to the active site. The interactions predicted involved hydrogen, hydrophobic, polar, and cation π bonds. However, the most active peptide, 108, did not meet the structural features proposed by Nongonierma and Fitzgerald.91

An in silico study based on the BIOPEP database was applied to determine the mechanism of release of active peptides from oat protein; nine active peptides (FFG, IFFFL, PFL, WWK, WCY, FPIL, CPA, FLLA, and FEPL) were predicted, subsequently chemically synthesized and evaluated in vitro. The inhibitory activities of the peptides were poor.98 However, other authors working with animal proteins have obtained inhibitory peptides against DPP-IV using this approach.81 The hydrophobicity of the two amino acids located at the N-terminal side was positively correlated with the DPP-IV inhibitory potency of peptides.

In silico methodologies are necessary for developing potent hydrolysates and peptides. Knowledge of the structural requirements for potent DPP-IV inhibitory peptides will constitute the basis for discovering novel potent DPP-IV inhibitory peptides. In silico tools, when applied at the molecular level, only sometimes have a high predictive ability as they fail to achieve a perfect match between predicted and actual peptide release. This is likely due to incorrect assumptions by peptide cutters which (a) only consider the primary sequence of proteins, while most proteins possess a secondary and tertiary structure, and (b) do not consider enzyme selectivity for peptide bond cleavage.99 However, to date, there need to be more human intervention studies that have demonstrated whether the ingestion of specific potent DPP-IV inhibitory hydrolysates induces an antidiabetic effect. These studies are necessary to (a) understand the relevance of the current research to human health and (b) determine effective hydrolysate doses required to observe an antidiabetic effect in vivo.

3.4.3. Terpenoids, alkaloids, chromenes, and furan derivatives. The terpenoids assayed included sesquiterpenoids and triterpenoids. These compounds were generally less active than flavonoids, and no kinetic analysis has been performed. The active compounds are listed in Table 3,100–109 and so far, boswelic acid derivative (151) showed the best activity level among terpenoids.
Table 3 Representative terpenoids and steroids with DPP-IV inhibitory activitya
Compound IC50 (μM) or inhibition (%) Control, IC50 (μM) or inhibition (%) Source Ref.
a IPI: diprotin A; SG: sitagliptin; BBR: berberine; NM: not mentioned.
11-Keto-boswellic acid (151) 1.7 SG, 0.2 Boswellia sacra Flueck (Burseraceae) 100
β-Boswellic acid (152) 3.1
1,2,6,10-Tetrahydroxy-3,9-epoxy-14-nor-5 (15)-eudesmane (153) 74.8 SG, 1.4 Flammulina velutipes (Curtis) Singer (Physalacriaceae) 101
1-Acetoxyalgoane (154) 43.3 IPI, 2.9 Laurencia natalensis Kylin (Rhodomelaceae) 102
8-Deoxyalgoane (155) 58.5
Algoane (156) 23.9
6,11-Dihydroxy-12-methoxy-5,8,11,13-abietatetraen-7-one (157) 228.9 SG, 5.4; IPI, 2.3 Caryopteris incana (Thunb. ex Houtt.) Miq. (Lamiaceae) 103
7,13,14-Trihydroxy-4-cadinen-15-oic acid methyl ester (158) 80.5 SG, 1.4 F. velutipes (Curtis) Singer (Physalacriaceae) 101
Diosgenin (159) 12.8 SG, 6.1 Trillium govanianum Wall. ex Royle (Melanthiaceae) 104
Borassoside D (160) 60.8
Borassoside E (161) 53.4
Pennogenin triglycoside (162) 56.3
Pennogenin tetraglycoside (163) 67.9
Caryopincaolide C (164) 54.2 SG, 5.4; IPI 2.3 C. incana (Thunb. ex Houtt.) Miq. (Lamiaceae) 103
Caryopincaolide D (165) 222.9
Caryopincaolide L (166) 168.7
Flammuspirone C (167) 75.9 SG, 1.4 F. velutipes (Curtis) Singer (Physalacriaceae) 101
Flammuspirone D (168) 83.7
Flammuspirone E (169) 70.9
Flammuspirone H (170) 79.7
Govanoside B (171) 61.4 SG, 6.1 T. govanianum Wall. ex Royle (Melanthiaceae) 104
Lupeol (172) 31.6 BBR, 13.3 Fagonia cretica Schreib. (Zygophyllaceae) 105
Macrocarpal A (173) 30% at 500 μM IPI, 30% at 25 μM Eucalyptus globulus Labill. (Myrtaceae) 106
Macrocarpal B (174) 30% at 500 μM
Macrocarpal C (175) 90% at 50 μM
Oleuropein (176) 72.3 NM Olea europaea L. (Oleaceae) 107
Peimisine (177) 80.5 IPI, 58.0 Fritillaria cirrhosa D. Don (Liliaceae) 108
Prineoparaquinone (178) 115.9 SG, 5.4; IPI 2.3 C. incana (Thunb. ex Houtt.) Miq. (Lamiaceae) 103
Protodioscin (179) 17.7 SG, 6.1 T. govanianum Wall. ex Royle (Melanthiaceae) 104
Quinovic acid (180) 30.7 BBR, 13.3 F. cretica Schreib. (Zygophyllaceae) 105
Quinovic acid 3β-O-β-D-glycopyranoside (181) 57.9
Quinovic acid-3β-O-β-D-glucopyranosyl-(28→1)-β-D-glucopyranosyl ester (182) 23.5
Salvicanaraldehyde (183) 178.3 SG, 5.4; IPI, 2.3 C. incana (Thunb. ex Houtt.) Miq. (Lamiaceae) 103
Stellasterol (184) 427.4 SG, 0.7 Ganoderma austral (Pers.) Bres (Ganodermataceae) 109


Among the alkaloids (Table 4),110–112 ephedrine derivatives (185–190) exhibited IC50 values between 124 μM to 28 mM; these relatively high values show their lack of selectivity towards other DPP isoforms, such as DPP-8 and DPP-9. On the other hand, the most active are berberine (191) and palmitine (192); docking studies predicted that they bind to the catalytic triad of DPP-IV. Berberine (191) has been used as an adjuvant for TIIDM for a long time. According to Jugran et al.,74 several studies demonstrated that 191 controls the glucose and lipid metabolism by modulating AMPK, GLUT-4, and α-glucosidase activity, reduction of glycated hemoglobin (HBA1c), postprandial blood glucose, fasting blood glucose and plasma triglycerides.74

Table 4 Miscellaneous natural products with DPP-IV inhibitory activitya
Compound IC50 (μM) Control, IC50 (μM) Source Ref.
a IPI: diprotin A; SG: sitagliptin; VG: vildagliptin; NM; not mentioned; CT: commercial standard.
Alkaloids
(1R,2R)-(−)-Norpseudoephedrine (185) 5020.0 VG, 3500.0 Ephedra sp. (Ephedraceae) 110
(1R,2S)-(−)-Ephedrine (186) 124.0
(1R,2S)-(−)-N-Methylephedrine (187) 27[thin space (1/6-em)]850.0
(1S,2R)-(+)-Norephedrine (188) 132.0
(1S,2S)-(+)-Pseudoephedrine (189) 4974.0
(1S,2S)-(+)-N-Methylpseudoephedrine (190) 28[thin space (1/6-em)]890.0
Berberine (191) 16.3 NM Coptis chinensis Franch (Ranunculaceae) 111
13.3 SG, 1.1 CT 112
Palmatine (192) 8.7
[thin space (1/6-em)]
Cromenes
11-(3,4,4a,5,8,8a-Hexahydro-8-methoxy-4-methyl-1H-isochromen-4-yloxy)-11-hydroxyethyl pentanoate (193) 467.6 IPI, 0.1 Sepiella inermis, Van Hasselt (Sepiidae) 113
Methyl 9-(4,4a,5,8-tetrahydro-3-oxo-3H-isochromen-5-yl) hexanoate (194) 826.9
[thin space (1/6-em)]
Furan derivatives
5-(7-(5-Ethyl-3,4-dimethoxycyclooctyl) benzofuran-6-yl)-7-methyl-3,4,7,8-tetrahydro-2H-oxocin-2-one (195) 2.0 IPI, 3.0 Gracilaria opuntia Durairatnam (Gracilariaceae) (https://www.marinespecies.org/aphia.php?p=taxdetails%26id=143748) 114
2-(3- Ethyl-9-(2-methoxyethoxy)-1-oxo-tetrahydro-1H-xanthen-2-yl) ethyl-5-hydroxy-9-methoxy-6,8-dimethyl-8-(5-methylfuran-2-yl) nona-3,6- dienoate (196) 2.0


The only two chromenes (193 and 194)113 tested using fluorimetric assays were less active than the positive control IPI; the authors indicated that the docking studies predicted their binding to the active site. However, their prediction was not confirmed using enzyme kinetic studies. Finally, two furanyl derivatives (195 and 196) isolated from Gracilaria opuntia were very active as DPP-IV inhibitors (IC50 2.0 μM, for both); their activities were comparable with the positive control (IPI, IC50 3.0 μM).114

image file: d3np00007a-u9.tif

image file: d3np00007a-u10.tif

image file: d3np00007a-u11.tif

image file: d3np00007a-u12.tif

image file: d3np00007a-u13.tif

image file: d3np00007a-u14.tif

image file: d3np00007a-u15.tif

image file: d3np00007a-u16.tif

image file: d3np00007a-u17.tif

image file: d3np00007a-u18.tif

image file: d3np00007a-u19.tif

3.4.4. Patents on natural DPP-IV inhibitors. More than twenty patents dealing with natural products have been reported and recently reviewed by Costante and Hussain.115,116 Their value is related more to the possible application in nutraceutical and functional foods as a dietary supplement, than as real drugs. These compounds include flavonoids, peptides, terpenoids, and alkaloids.

4. Diacylglycerol acyltransferase inhibitors

4.1. Structure of DGAT

Diacylglycerol acyltransferase (DGAT), an endoplasmic reticulum (ER)-membrane-bound enzyme, belongs to the membrane-bound O-acyltransferase (MBOAT) family. DGAT catalyzes the conversion from diacylglycerol (DAG) to triacylglycerol (TAG) in the glycerol phosphate pathway (Fig. 4). This enzyme has two isoforms, DGAT-1 and DGAT-2, present in white adipose tissue. DGAT-1 is mainly found in the small intestine, and DGAT-2 in the liver. The selective inhibition of DGAT-1 is actively investigated due to its potential for treating obesity and TIIDM. DGAT-2 may be a target for treating DGAT-2-related liver diseases, e.g., hepatic steatosis, hepatic injury, and fibrosis.117
image file: d3np00007a-f4.tif
Fig. 4 DGAT catalyzes the final committed step in the biosynthesis of triglycerides.

DGAT isoforms are encoded by two different genes widely expressed in mammals. In humans, the DGAT-1 gene, found on chromosome 8, has 10.62 kb and 17 exons. This gene encodes a protein of ∼500 residues in most species with a calculated molecular mass of ∼55 kDa. The Dgat2 gene encodes a ∼42 kDa protein with a ‘hairpin-like’ structure. To date, the crystal structure of DGAT-2 has not been reported. The structure and catalytic mechanism of DGAT-1 remained unknown until 2020, when Sui et al.118 determined the structure of dimeric human DGAT-1 by cryo-electron microscopy with a resolution of approximately 3.0 Å (PDB code: 6VYI). DGAT-1 forms a homodimer through N-terminal segments and a hydrophobic interface. Its active sites are located within the membrane region. The protein has nine transmembrane helices, eight forming MBOAT fold structure. The crystal structure of DGAT-1 complexed with the oleoyl-CoA substrate (PDB code: 6VZ1; Fig. 5) shows that the CoA moiety binds DGAT-1 on the cytosolic side. The acyl group is in a hydrophobic channel, leaving the acyl-CoA thioester bond near a conserved catalytic histidine residue.119 The highly conserved catalytic residues exposed laterally to the membrane bilayer allow lipid access to the active site.


image file: d3np00007a-f5.tif
Fig. 5 Structure of human DGAT-1 complexed with acyl-CoA substrate by Cryo-EM. Ribbon representation of the human DGAT-1 dimer. The dashed line indicates a disordered segment (residues 229–238) not resolved in the cryo-EM map (https://www.rcsb.org/structure/6VZ1).

As mentioned above, we will focus on reviewing selective inhibitors of DGAT-1 as a potential target for treating obesity and type II diabetes. Several DGAT-1 inhibitors with promising therapeutic potential for obesity (Fig. 6) have gone into clinical trials, for example, LCQ-908 (Novartis), AZD-7687 (AstraZeneca), and PF-04620110 (Pfizer). Unfortunately, none has successfully concluded the clinical trials, some due to adverse skin and adipose tissue effects.117


image file: d3np00007a-f6.tif
Fig. 6 Synthetic DGAT-1 inhibitors developed by pharmaceutical companies.

4.2. Assay methods

DGAT-1 is ubiquitously expressed; however, its remarkably high expression in adipocytes and small intestine enterocytes indicates that this enzyme has a significant role in lipid metabolism. In adipocytes, DGAT-1 catalyzes the formation of TAG via the condensation of DAG and a range of fatty acids (FA). Accordingly, the synthesis of TAG should be quantitatively affected by the inhibition of this enzyme in adipocytes. Moreover, TAG synthesis should also decrease due to the inhibition of DGAT-1 in enterocytes. This, in turn, would affect TAG levels in the systemic circulation following an oral lipid load because of the following. FA and monoacylglycerol in the GI tract are absorbed by enterocytes and are then condensed into TAG. This is crucial for the assembly and secretion of chylomicrons that transport TAG into the bloodstream for further use by high-energy-requiring tissues, such as muscle.117

There are several animal models to assess DGAT inhibition by small molecules (adipose TAG synthesis in rats, oral lipid tolerance in rats, diet-induced obesity mice, etc.).120 Yet, the rational discovery of potential inhibitors is often achieved by a combination of in vitro assays and in silico methods (docking, pharmacophore, 3D-QSAR models, machine learning methods, etc.).117,121

HepG2 cells are used for in vitro assays because lipoprotein assembly heavily relies on high concentrations of exogenous fatty acids in these cells. DGAT inhibition is measured by the secretion of apolipoprotein B (apoB) in cells incubated with oleic acid (OA) and the test compound.117 Other methods use microsomes from rat liver. In both cases, whole-cell esterification of DAG is then quantified by adding [14C]palmitoyl-CoA and measuring the [14C]TAG formed by TLC or liquid scintillation counting.117

An in vitro assay for human DGAT-1 (hDGAT-1) inhibitors involves the expression of the enzyme using a baculovirus expression system in Sf9 insect cells.122 Once the enzyme is extracted, serial dilutions of the tested compounds and [14C]oleoyl coenzyme A are added to the reaction mixture in a 96-well plate. The lipids are then separated, and the hDGAT-1 activity is quantified by counting aliquots of the upper heptane layer by liquid scintillography.117 Finally, an in vitro and in vivo DGAT-selective assay to discern between the roles of DGAT-1 and DGAT-2 in TAG synthesis uses isotope-labeled [13C18] oleic acid. Then LC-MS-MS analysis is used to measure the [13C18] oleoyl incorporated into TAG and DAG.123

4.3. Natural products with DGAT-1 inhibitory properties

Many natural products from plants and microorganisms, have been reported to inhibit DGAT activity (Tables 5 and 6). Like DPP-IV inhibitors, the largest group of DGAT inhibitors are flavonoids and other polyphenols (Table 5). For example, the flavonoids isoliquilitin (197), liquiritigenin (201), isoliquiritigenin (16), liquilitin (202), and glabrol (203) were isolated from the roots of Glycyrrhiza uralensis Fisch. (Fabaceae) and tested in an in vitro DGAT inhibitory assay. Glabrol (203) showed non-competitive inhibition against DGAT with an IC50 value of 8 μM. The use of licorice roots for treating obesity and TIIDM patients could be related to the DGAT inhibitory activity of this flavonoid.124
Table 5 Representative natural products from plants with DGAT inhibitory activitya
Compound IC50 (μM) or inhibition (%) Control, IC50 (μM) Source Ref.
a KD: kuraridin; CPT: cryptotanshinone; EGCG: epigallocatechin gallate; BA: betulinic acid; EC: evocarpine; NM; not mentioned; CT: commercial standard.
Chalcones
Isoliquiritigenin (16) >300.0 KD, 10.2 Glycyrrhiza uralensis Fisch. (Fabaceae) 124
Isoliquiritin (197) >300.0
Xanthohumol (198) 50.3 NM Humulus lupulus L. (Cannabaceae) 127
Xanthohumol B (199) 194.0
Kuraridin (200) 9.8 CPT, 35.5 Sophora flavescens Aiton (Fabaceae) 125
[thin space (1/6-em)]
Flavanones
Liquiritigenin (201) 195.2 KD, 10.2 G. uralensis Fisch. (Fabaceae) 124
Liquiritin (202) 110.6
Glabrol (203) 8.0
Kurarinone (204) 10.9 CPT, 35.5 S. flavescens Aiton (Fabaceae) 125
Kurarinol (205) 8.6
Kushenol H (206) 142.0
Kushenol K (207) 250.0
2(S)-4′-Hydroxy-6-methoxy-7-(2′′-hydroxy-3′′-methybult-3′′-enyl) flavanone (208) 70.4 KD, 10.3 Psoralea corylifolia (L.) Medik. (Fabaceae) 140
2(S)-4′-Hydroxy-7-methoxy-6-(1′′,2′′-epoxy-3′′-hydroxy-dimethyl) flavanone (209) 89.2
[thin space (1/6-em)]
Flavanonols
Taxifolin (23) 35% at 200 μM NM CT 141
[thin space (1/6-em)]
Isoflavones
8-Prenylleutone (210) 35.5 KD, 11.0 Erythrina senegalensis D.C. (Fabaceae) 126
Auriculatin (211) 17.1
Erysenegalensein O (212) 2.5
Erysenegalensein D (213) 3.4
Erysenegalensein N (214) 28% at 12.5 μM
Derrone (215) 44.9
Alpinumisoflavone (216) 23% at 12.5 μM
6,8-Diprenylgenistein (217) 16.5
Hydroxypsoralenol A (218) >250.0 KD, 10.6 P. corylifolia (L.) Medik. (Fabaceae) 135
Hydroxypsoralenol B (219) >250.0
Psoralenol (220) >250.0
[thin space (1/6-em)]
Flavonol
Quercetin (68) 48% at 15 μM NM CT 142
[thin space (1/6-em)]
Polyphenols
Rugosin B (221) 96% at 10 μM EGCG (inactive) Rosa centifolia L. (Rosaceae) 129
Rugosin D (222) 82% at 10 μM
Eusupinin A (223) 84% at 10 μM
Bavacoumestan C (224) >250.0 KD, 10.6 P. corylifolia (L.) Medik. (Fabaceae) 135
Bavacoumestan B (225) >250.0
7,2′,5′-Trihydroxy-8-prenylaurone (226) 52.2 KD, 10.3 P. corylifolia (L.) Medik. (Fabaceae) 140
Damaurone C (227) >200.0
Damaurone D (228) >200.0
(Z)-2-(4-Methoxybenzylidene)-7,7-Dimethyl-7,8-dihydro-2H-furo [2,3-f]chromene-3,9-dione (229) >200.0
(Z)-4,6-Dimethoxy-7,4′-dihydroxyaurone (230) >200.0
Ruguarone B (231) >200.0 KD, 10.3 P. corylifolia (L.) Medik. (Fabaceae) 140
6,7,3′,4′-Tetrahydroxyaurone (232) >200.0
Licoagroaurone (233) 54.4
Glyinflain A (234) 58.6
[thin space (1/6-em)]
Terpenoids
Brachynereolide (235) >250.0 CPT, 27.3 Youngia koidzumiana Kitam. (Asteraceae) 132
Ixerin Y (236) >250.0
Crepidiaside C (237) >250.0
Cryptotanshinone (238) 35.5 NM Salvia miltiorrhiza Bunge (Lamiaceae) 131
Tanshinone IIA (239) >850.0
15,16-Dihydrotanshinone I (240) 39.9
Tanshinone I (241) >850.0
Aphadilactone A (242) 26% at 10 μM BA, 17.2 Aphanamixis grandifolia Blume (Meliaceae) 133
Aphadilactone B (243) 9% at 10 μM
Aphadilactone C (244) 0.5
Aphadilactone D (245) 14% at 10 μM
Echinocystic acid (246) 138.0 NM Gleditsia sinensis Lam. (Fabaceae) 134
Germanicol acetate (247) >250.0 CPT, 27.3 Y. koidzumiana Kitam. (Asteraceae) 132
Oleanolic acid (248) 31.7
Methyl ursolate (249) 26.4 CPT, 27.3 Y. koidzumiana Kitam. (Asteraceae) 132
Corosolic acid (250) 443.0
12α-Psoracorylifol F (251) 71.7 KD, 10.6 P. corylifolia (L.) Medik. (Fabaceae) 135
7β,8α-Hydroxy-12 β-psoracorylifol F (252) 76.1
8-Ketone-cyclobakuchiol C (253) 89.1
7α,8β-Hydroxy-12 β-cyclobakuchiol C (254) 61.5
8α-Hydroxy-cyclobakuchiol C (255) 67.7
Psoracorylifol F (256) 75.4
[thin space (1/6-em)]
Alkaloids
1-Methyl-2-tetradecyl-4(1H)-quinolone (257) 69.5 NM E. rutaecarpa (A. Juss.) Benth. (Rutaceae) 130
Evocarpine (258) 23.8
1-Methyl-2-[(4Z,7Z)-4,7-decadienyl]-4(1H)-quinolone (259) 20.1
1-Methyl-2-[(6Z,9Z)-6,9-pentadecadienyl]-4(1H)-quinolone (260) 13.5
Retrofractamide C (261) >900.0 KD, 9.8 P. longum L. (Piperaceae) and P. longum L. (Piperaceae) 143
(2E,4Z,8E)-N-[9-(3,4-Methylenedioxyphenyl)-2,4,8-nonatrienoyl]-piperidine (262) 29.8
Pipernonaline (263) 37.2
Piperrolein B (264) 20.1
Dehydropipernonaline (265) 21.2
[thin space (1/6-em)]
Others
(9R,10S)-Epoxyheptadecan-4,6-diyn-3-one (266) 34.6 EC, 23.9 Panax ginseng C.A. Mey. (Araliaceae) 144
1-Methoxy-(9R,10S)-epoxyheptadecan-4,6-diyn-3-one (267) 110.3


The prenylflavonoids, kurarinone (204), kurarinol (205), kushenols H (206) and K (207), and the chalcone kuraridin (200), isolated from the roots of Sophora flavescens Aiton (Fabaceae), inhibited DGAT activity in a concentration-dependent manner with IC50 values of 10.9, 8.6, 142, 250, and 9.8 μM. Interestingly, compounds without C3-OH (200, 204, and 205) showed more potent inhibition than those with C3-OH (206, and 207).125 In addition, kurarinone (204) also inhibited the de novo synthesis of TAG in Raji cells. Compound 200 has been used as a positive control in many in vitro studies.

A bioassay-guided fractionation of an extract of the stem bark of Erythrina senegalensis led to the isolation of a series of prenylflavonoids (210–217). From these, 8-prenylleutone (210), auriculatin (211), erysenegalensein O (212), erysenegalensein D (213), derrone (215), and 6,8-diprenylgenistein (217) inhibited DGAT activity with IC50 values ranging from 1.1 to 15.1 μg mL−1.126

The chalcone xanthohumol (198) was also described as an inhibitor of DGAT.127 Compound 198 is a potent inhibitor of apoB secretion and enhances apoB degradation. It was also reported that xanthohumol (198) decreases the expression of DGAT-1 mRNA, has hypoglycemic effects, and decreases hepatic triglyceride in KK-Ay mice.128

image file: d3np00007a-u20.tif

image file: d3np00007a-u21.tif

image file: d3np00007a-u22.tif

image file: d3np00007a-u23.tif

The ellagitannins rugosins B (221) and D (222) and eusupinin A (223) isolated from rose petals inhibited the DGAT activity by 96%, 82%, and 84%, respectively, at 10 μM.129 Remarkably, the activities of other microsomal enzymes were not affected.

image file: d3np00007a-u24.tif

image file: d3np00007a-u25.tif

image file: d3np00007a-u26.tif

Plant alkaloids have also shown important DGAT inhibitory activity as demonstrated for the quinolone alkaloids 257–265 isolated from the fruit of Evodia rutecarpa. The evocarpine derivates 258–260 showed concentration-dependent inhibition with IC50 values below 24 μM.130 Other examples are the alkamides 261–265 isolated from the fruits of Piper longum and P. nigrum (Piperaceae). From these, piperrolein B (264) was the most potent, with an IC50 value of 20.1 μM.

Among the plant terpenoids active against the activity of DGAT,131–135 the dimeric diterpenoid, aphadilactone C (245), isolated from the leaves of Aphanamixis grandifolia (Meliaceae), is the most potent natural DGAT-1 inhibitor discovered to date (IC50 = 0.46 μM, selectivity index > 217 against DGAT-2).133A. grandifolia, is a tree that grows in Asia, and its leaves and roots are used in traditional Chinese medicine. In addition, a SAR analysis of diastereoisomers 242–245 revealed that the active binding site of the DGAT-1 enzyme is very selective to the stereochemistry of the substrate.133 Therefore, compound 245 could lead to the development of potent and selective DGAT-1 inhibitors.

image file: d3np00007a-u27.tif

image file: d3np00007a-u28.tif

image file: d3np00007a-u29.tif

A series of polyacetylenes, diterpenes, tridepsides, fatty acid derivatives, and phenylpyropenes from algae136 and fungi have shown DGAT inhibitory activity (Table 6). Particularly, from a series of amidepsines, roselipins, and phenylpyropenes, isolated from the fungal strains Humicola sp. FO-2942 and FO-5969 (soil isolates), Gliocladium roseum KF-1040 (marine isolate), and Penicillium griseofulvum F1959 (soil isolate),137–139 amidepsine A (270, IC50 = 10.2 μM) and phenylpyropene C (280, IC50 = 11.04 μM) were the most potent inhibitors, with a potency comparable to kuraridin (200). Compound 280 showed an inhibitory effect in rat liver microsomes (IC50 = 10.2 μM) and Raji cells (IC50 = 15.5 μM) and is commercially available as a standard for the screening of DPP-IV inhibitors. While amidepsines B (271) and D (273) showed specific inhibition on TAG formation in Raji cells.137 In the group of the phenylpyropenes, 280 showed a noncompetitive pattern of enzyme inhibition.139

image file: d3np00007a-u30.tif

image file: d3np00007a-u31.tif

image file: d3np00007a-u32.tif

image file: d3np00007a-u33.tif

Table 6 Representative natural products from chromista and fungal origin with DGAT inhibitory activitya
Compound IC50 (μM) Control, IC50 (μM) Source Ref.
a KD: kuraridin; XH: xanthohumol; NM; not mentioned.
Diterpenes from chromista origin
Dictyol E (268) 46.0 XH, 50.3 Pachydictyon coriaceum (Holmes) Okamura (Dictyotaceae) 136
Hydroxyisocrenulatin (269) 23.3
[thin space (1/6-em)]
Miscellaneous compounds from fungal origin
Amidepsine A (270) 10.2 NM Humicola sp. Traaen (Chaetomiaceae) FO-2942 and FO-5969 137
Amidepsine B (271) 19.2
Amidepsine C (272) 51.6
Amidepsine D (273) 17.5
Roselipins 1A (274) 17.0 NM Gliocladium roseum Corda (Hypocreaceae) KF-1040 138
Roselipins 1B (275) 15.0
Roselipins 2A (276) 22.0
Roselipins 2B (277) 18.0
Phenylpyropenes A (278) 78.7 KD, 9.8 Penicillium griseofulvum Dierckx R.P. (Trichocomaceae) F1959 139
Phenylpyropenes B (279) 21.7
Phenylpyropenes C (280) 11.0
Isochaetochromin A1 (281) 320.0 NM Penicillium sp. (Trichocomaceae) FKI-4942 145
Isochaetochromin B1 (282) 190.0
Isochaetochromin B2 (283) 310.0


5. Fructose 1,6-biphosphatase inhibitors

5.1. Structure of FBPase

Glycogenolysis and gluconeogenesis (GNG) are the main pathways for glucose formation in the human liver. The latter, where three-carbon precursors such as pyruvate, lactate, and glycerol are converted into glucose, is the main process for glucose output in a fasting state: over 50% of glucose production is formed via GNG during persistent fasting or starvation; this is even higher in TIIDM patients.146 The GNG pathway involves the conversion of fructose 1,6-bisphosphate (FBP) into fructose 6-phosphate (F6P), which is then converted into glucose after catalysis by glucose 6-phosphate isomerase and glucose 6-phosphatase. The reaction from FBP to F6P is catalyzed by fructose 1,6-bisphosphatase (FBPase), a rate-limiting enzyme (Fig. 7). In mammals, two genes encode FBPase: FBP1, mainly found in the liver and kidney, and FBP2, found exclusively in muscle tissue.147 FBP1 and FBP2 overall identity is ∼77%, and the regions involved in binding the substrate, regulatory molecules, and magnesium are almost identical. The physiological role of muscle FBPase remains unclear because muscle tissue has no glucose 6-phosphatase activity and is thus nongluconeogenic. Probably, muscle FBPase is needed for glycogenesis from lactate. The liver form of FBPase regulates blood glucose levels in the liver and kidney. Compared to the liver, the kidney is a minor source of de novo synthesized glucose except during extreme fasting, and, reportedly, in a diabetic state.147 In this context, human liver FBPase has been considered a potential therapeutic target for the treatment of TIIDM. In patients and animal models of TIIDM, the expression levels and activities of FBPase were upregulated in the liver, indicating the importance of this enzyme for increasing blood glucose.
image file: d3np00007a-f7.tif
Fig. 7 FBPase catalyzes the conversion of FBP into F6P and then into glucose during GNG.

FBPase is a cytosolic homotetramer, where each monomer (36.7 kD) presents a substrate (FBP) binding site and an AMP allosteric regulatory site within 28 Å of the active site (Fig. 8).148 The enzyme has two conformational states, active (R) and inactive (T). A conformational change from R to T or the stabilization of T result from the allosteric inhibition of AMP. Another allosteric binding site was recently found for the inhibitor OC252.149 Also, the enzyme undergoes competitive substrate inhibition by fructose 2,6-bisphosphate.


image file: d3np00007a-f8.tif
Fig. 8 Structure of human liver FBPase complex with Mg2+ and AMP (https://www.rcsb.org/structure/7C9Q).

The development of FBPase inhibitors as potential antidiabetic agents remains a challenging task. The difficulties arise from the complex hydrophobic region on the subunit interface of FBPase and the highly charged substrate-binding site. Potent and selective competitive and uncompetitive inhibitors are thus scarce.147 Pharmaceutical companies and academic laboratories have investigated a wide array of noncompetitive (allosteric) inhibitors of the AMP binding site in FBPase (Fig. 9).150 Some synthetic FBPase inhibitors effectively decrease hepatic glucose production and blood glucose levels in diabetic animal models. However, only AMP mimetic MB07803, a phosphonic acid-containing thiazole prodrug of MB07729, has reached phase II in clinical trials.


image file: d3np00007a-f9.tif
Fig. 9 Synthetic FBPase inhibitors.

5.2. Assay methods

Since its original isolation from rabbit liver and kidney in 1943,151 the activity of FBPase has been assayed (a) spectrophotometrically by following the triphosphopyridine nucleotide (TNP) reduction or the reduction of NADP+ to NADPH in the presence of glucose 6-phosphate isomerase and glucose 6-phosphate dehydrogenase,151–153 or by the release of inorganic phosphate (Pi) from FBPase using the Malachite Green assay152 or by the of Fiske and Subbarow154 or Tausky and Shorr155 methods; (b) radiometrically by measuring the release of 32Pi from fructose l,6-[l-32P]P2;156 and (c) by LC-ESI-MS analysis.157

In addition, FBPase from pig kidneys and sheep liver have been purified and used for assays, and their complete amino acid sequences have been determined.158 The homology of all mammal enzyme forms extends to those from plants, yeast, and Escherichia coli, indicating that FBPase has undergone minimal changes throughout evolution, even though regulation of the enzyme's activity differs in different cell types. In 1993, the isolation of the cDNA for the human liver enzyme, the construction of a plasmid for its expression in E. coli, and purification and characterization of the recombinant enzyme was reported, as well as the expression of mutant enzymes by site-directed mutagenesis of active-site residues.158 The human enzyme has been used in the same protocols mentioned for the rabbit FBPase.159–161 Recently, a new assay, the PicoProb™ FBPase inhibitor screening kit by BioVision, Inc., was developed to discover novel inhibitors. Briefly, human FBPase hydrolyzes fructose 1,6-bisphosphate to fructose 6-phosphate, which oxidizes and reacts with a fluorogenic probe that is detected at λEm/Ex = 535/587 nm.162

5.3. Natural products with FBPase inhibitory properties

Several synthetic FBPase inhibitors bearing distinct scaffolds have been identified via either high-throughput screening of compound libraries or structure-guided design of AMP mimetics.162 These include: (1) uncompetitive inhibitors (anilinoquinazoline derivatives, pseudo-tetrapeptide, and quinoline derivatives); (2) non-competitive phosphorous based inhibitors (purine derivatives, benzimidazole derivatives, thiazole derivatives, and oxazole derivatives); and (3) other non-competitive inhibitors (indole derivatives, benzoxazole derivatives, various N, O, S containing heterocycles derivatives, sulfonyl derivatives, and phytochemicals). Among these, only the AMP mimetic MB07803 has advanced to phase II clinical trials. In contrast, natural products with FBPase inhibitory activity have been barely studied (Table 7). In the following lines, we will discuss the most active natural inhibitors discovered to date.
Table 7 Representative natural products with FBPase inhibitory activitya
Compound IC50 (μM) or inhibition (%) Control, IC50 (μM) or inhibition (%) Source Ref.
a a Liver, b kidney, and c muscle FBPase from diabetic rats. FBPase activity measured as μmoles of d Pi liberated per min per mg protein or e Pi liberated per hour per mg protein in the presence of f 100, g 50, h 40, i 25, j 20 or k 10 mg per kg of the compound. l Human liver and m pig kidney FBPases inhibition in the presence of n 200, o 10, and p 1 μM of the compound.
Flavanone
Eriodictyol (19) <20%l,n AMP, 2.6 μMl CT 162
Hesperitin (284) 37%a,e,h NM CT 163
[thin space (1/6-em)]
Flavanonol
Taxifolin (23) <20%l,n AMP, 2.6 μMl CT 162
[thin space (1/6-em)]
Flavone
Naringenin (21) <20%l,n; 13%l,o; 5%l,p AMP, 2.6 μMl; 91%l,o; 26%l,p; 2.9 μMl CT 162
Chrysin (25) <20%l,n AMP, 2.6 μMl CT
Baicalein (27) 29.0l CT
Acacetin (28) <20%l,n CT
Apigenin (29) <20%l,n; 15%l,o; 15%l,p AMP, 2.6 μMl; 91%l,o; 26%l,p; 2.9 μMl CT
Luteolin (39) 22%l,n AMP, 2.6 μM l CT
Galangin (47) <20%l,n CT
Myricetin (65) <20%l,n CT
Diosmin (285) 38%a,e,f NM CT 164
2-Phenylchromone (286) <20%l,n AMP, 2.6 μMl CT 162
Baicalein 7-methyl ether (287) 46%l,n CT
Baicalein trimethyl ether (288) <20%l,n CT
Baicalin (289) <20%l,n CT
Scutellarein (290) 38.2 μMl CT
Quercetagenin (291) 37%l,n CT
Norwogonin (292) <20%l,n CT
Gossypetin (293) 24.0 μMl CT
Gossypin (294) <20%l,n CT
8-Prenylquercetin (295) 87%l,o; 19%l,p; 3.6 μMl AMP, 91%l,o; 26%l,p; 2.9 μMl Desmodium caudatum (Thunb.) DC. (Fabaceae) 165
[thin space (1/6-em)]
Anthocyanin
Gossypetinidin chloride (296) 46%l,n AMP, 2.6 μMl CT 162
[thin space (1/6-em)]
Isoflavone
Biochanin A (297) 33.3%a,e,k NM CT 166
21.9%b,e,k
[thin space (1/6-em)]
Flavonol
Chrysoeriol (26) <20%l,n AMP, 2.6l CT 162
Morin (50) 14%a,e,i; 29%a,e,g; <20%l,n CT 162 and 167
Rutin (51) 32%a,d,f; 25%b,d,f; 31%c,d,f; <20%l,n CT 162 and 168
Kaempferol (56) <20%l,n CT 162
Quercetin (68) 26%a,e,j; 38%b,e,f; 28%c,e,f; <20%l,n; 25%l,o AMP, 2.6 μMl; 91%l,o; 26%l,p; 2.9 μMl CT 162, 165 and 169
Herbacetin (298) 8.7 μMl AMP, 2.6 μMl CT 162
[thin space (1/6-em)]
Benzofurane
(+)-Usnic acid (299) 371.0 μMl; 930.0 μMm AMP, 9.8 μMl; 1.3 μMm CT 170


Significant FBPase inhibition was reported for the flavonoids, baicalein (27), scutellarein (290), herbacetin (298), and gossypetin (293), with mean IC50 = 29, 38.2, 8.7, and 24 μM, respectively. The positive control, AMP, exhibited an IC50 value of 2.6 μM.162 Herbacetin (298), a natural flavonoid containing –OH groups at C-3 of ring C, at C-4′ of ring B, and at C-5, C-7, and C-8 of ring A, is found in flaxseed and other plants. This natural product also shows antihyperglycemic and antihyperlipidemic activities thus protecting against the effects of chronic high-fat diet consumption (associated with insulin resistance).162 Moreover, the potential of 298 for the management of TIIDM has been acknowledged because of its interaction with aldose reductase and the insulin receptor. Interestingly, ingesting flaxseed oil or flaxseed supplementation has proven to reduce glucose levels and glycosylated hemoglobin concentration and increase insulin sensitivity in humans.162

Antitumor, anti-inflammatory, antioxidant, and antidiabetic properties have been reported for baicalein (27),162 a component of the Chinese plant Scutellaria baicalensis. Scutellarein (290), found in the perennial herb Scutellaria lateriflora, is the hydrolyzed, and highly soluble form of scutellarin. Scutellarin has been used in China since 1984 to treat acute cerebral infarction and paralysis induced by cerebrovascular diseases. Scutellarin is mainly absorbed in the form of 290, which is likely the actual bioactive component.162 Gossypetin (293), is usually found in the flowers and the calyces of Hibiscus sabdariffa. This plant has traditionally been used as a sedative and for treating nervous disorders. In addition to its anxiolytic and antidepressant activities, this flavonoid is associated with antimicrobial, antiatherosclerotic, and anti-inflammatory effects.162

The flavonoid 8-prenylquercetin (296) showed potent α-glucosidase and FBPase dual inhibitory activity, with IC50 values of 4.38 and 3.62 μM, respectively.162

Finally, (+)-usnic acid (299), a naturally occurring dibenzofuran derivative found in several epiphytic lichen species, inhibits both human liver and pig kidney FBPases in vitro with IC50 values of 0.37 mM and 0.93 mM, respectively.170

image file: d3np00007a-u34.tif

image file: d3np00007a-u35.tif

6. Glucokinase activators

6.1. Structure of GK

Glucokinase (GK) is a 52 kDa monomeric enzyme comprising 465 residues arranged in two domains, a large and a small α/β domain, separated by a deep cleft that functions as the active site for phosphorylation. The small domain comprises of residues 66–202, and helix C (or C-terminus) contains residues 442–465. The large domain encloses the rest of the protein (Fig. 10).171 Crystallographic analysis of GK structure has revealed that this enzyme has three conformations: super-open, open, and closed.172 The super-open conformation is characterized by an opening angle of 100° between the large and small domains and a disordered loop formed by residues 151–180. In this state, the small domain residues are disorganized and unable to bind glucose. Thus, the super-open conformation predominates at low glucose concentration.173,174 In the absence of either glucose or activator, GK adopts a “super-open”, inactive apo-conformation. The closed conformation, the most compact, has an opening angle of 40°. Glucose phosphorylation is then carried out in the presence of ATP. This process is facilitated by Lys169 residue, which acts as an acid catalyst and enhances the binding of GK with both ATP and glucose.175 Once the phosphorylation is completed, GK shifts to the open conformation to release catalytic product, G6P (and ADP), and slowly equilibrates to the super-open conformation. The catalytic cycle re-starts if glucose binds to the open conformation (fast cycle) and slows if glucose concentrations decrease (slow cycle) as this state favors super-open conformation, thus explaining the sigmoidal saturation curve for glucose of GK.173
image file: d3np00007a-f10.tif
Fig. 10 Ribbon representation of the human GK complexed with a benzamide derivative, α-D-glucopyranose, and Na+ (https://www.rcsb.org/structure/1V4S).

6.2. GK and glucose homeostasis

After carbohydrate ingestion and hydrolysis in the gastrointestinal tract, dietary monosaccharides are transferred from the luminal intestinal surface across cellular membranes and then transported to other cells. Monosaccharides are phosphorylated by ATP-dependent phosphorylating enzymes known as kinases which are stored or used as substrates for cellular metabolic processes. Hexokinases (EC 2.7.1.1) phosphorylate hexoses such as 2-deoxyglucose, mannose, glucosamine, fructose, and glucose, usually at the hydroxyl at the 6′-position in a six-carbon sugar, using ATP as the phosphate donor (Fig. 11). Four hexokinase isoforms have been identified in mammals: hexokinase 1 (HK1, I or A), hexokinase 2 (HK2, II or B), hexokinase 3 (HK3, III or C), and hexokinase 4 or glucokinase (HK4, IV, D or GK).174,176 GK is not inhibited by G6P, and its concentration is proportional to the extracellular glucose levels. Accordingly, GK activity can be regarded as a glucose sensor in several tissues, particularly those involved in glucose homeostasis, such as the liver and pancreas.176–178 For instance, in hepatocytes, GK promotes glycogen synthesis, particularly during the postprandial state. On the other hand, in pancreatic β cells, GK regulates the entry of glucose into the glycolytic pathway and its subsequent metabolism, therefore controlling glucose-dependent insulin secretion.179–186.
image file: d3np00007a-f11.tif
Fig. 11 Reaction mechanism of glucose phosphorylation catalyzed by GK.

The hyperglycemic state characteristic of DM is due to a combination of impaired glucose utilization and enhanced liver gluconeogenesis, both regulated by GK. Thus, GK activators have been considered an alternative to treat TIIDM. These pharmacological agents are predicted to have a dual effect when controlling glucose metabolism by increasing insulin secretion in pancreatic β-cells or stimulating glycolysis and glycogen synthesis in the liver.177 Development of synthetic GK activators have focused on identifying compounds that bind to allosteric sites in hepatic GK rather than in pancreatic β-cells GK, as in the latter case numerous early-developed GK activators induced severe hypoglycemia.171,179,187

6.3. Assay methods

Most of the experimental evidence on GK activation by any compound derives from sub-chronic in vivo experiments using tissues from rodents with either chemically- or diet-induced diabetes.188

Early GK activity determinations were carried out in cell cultures using D-(U-14C)-glucose and measured by scintillation counter. GK activity was proportional to radioactivity due to marked G6P.189 Nowadays, GK activity is assessed by using a NADPH-coupled assay with G6P dehydrogenase. The end product of the reaction is 6-P-gluconate and NADPH, which possesses an intrinsic fluorescence intensity; GK activity is proportional to the fluorescence intensity of NADPH.190 This procedure is useful for calculating the binding affinity constants (KD) and kinetic characteristics of GK activators.191 Another method is the luminescence assay, which monitors the ATP consumption associated with GK catalytic activity and uses a detection system based on luciferin and luciferase to produce a luminescent signal that is proportional to ATP. Therefore, the GK activity is inversely proportional to the ATP concentrations in the GK reaction.190 GK activity and expression is monitored by immunological and molecular biology techniques.192,193 Finally, hepatic mRNA expression of GK can be determined using quantitative real-time PCR, using appropriate primers for GK amplification.194,195

Since there are several methods to assess GK activation and GK protein expression, there are no consensus on units used to express the results, which makes it difficult to compare the efficacy of pure compound and extracts.

6.4. Plant products activators of GK

There are a few recent reports describing the effect of medicinal plant extracts on GK expression or activity using different assays in vitro or in vivo.181,196 These reports do not specify the mechanism of GK activation. Furthermore, GK hepatic activation data is accompanied by information on increased insulin secretion and/or glycogen synthesis. Many natural products can modulate GK activity (Table 8) or expression (Table 9). As in previously discussed enzymes, flavonoids are largest group of GK activators for by catechin (6) administered orally (20 mg kg−1 day−1 per 6 weeks) to STZ-diabetic rats also provoked an augmentation in insulin levels (8.6 ± 1.9 μmol mL−1) and liver glycogen (49.2 ± 5.3 mg per g tissue) compared to diabetic control (8.2 ± 1.4 μmol mL−1 and 21.5 ± 3.8 mg g−1, respectively).204 Likewise, flavanones hesperidin (20) and hesperetin (300) almost triplicated glycogen levels when given to STZ-diabetic rats (45.37 ± 3.14 mg per g tissue and 17.12 ± 1.31 mg per g tissue, respectively) compared to diabetic controls (26.19 ± 2.42 mg per g tissue and 10.75 ± 0.82 mg per g tissue, respectively) when administered either during 30 or 45 days at doses of 100 or 40 mg kg−1 day−1.207,208 When amentoflavone (301) was given to obese and diabetic mice (40 mg kg−1 day−1 per 8 weeks), it increased insulin secretion (21.39 ± 3.62 μU mL−1 respect to diabetic control, 16.12 ± 2.97 μU mL−1).202 In the same animal model, a daily dose of 100 mg kg−1 per 30 days of polymethoxylated flavone tangeretin-1 (302) significantly increased insulin (15.98 ± 1.49 μU mL−1) and liver glycogen levels (48.05 ± 4.14 mg per g tissue) compared to diabetic controls (6.26 ± 0.53 μU mL−1 and 15.77 ± 1.45 mg per g tissue, respectively).210 Similar effects were observed for the methoxylated isoflavone biochanin A (297) but at a lower dose (10 mg kg−1 day−1 per 45 days), with insulin and liver glycogen levels duplicated after treatment (13.34 ± 0.65 μU mL−1versus 7.55 ± 0.39 μU mL−1, and 45.56 ± 3.76 mg per g tissue versus 22.87 ± 2.18 mg per g tissue, respectively).203 Kaempferol (56, after 12 weeks of treatment, 50 mg kg−1 day−1) did not increase plasma insulin levels in diabetic mice but significantly augmented liver glycogen.209In silico studies predicted that flavonoids 301, 56, and 68 are allosteric GK activators.235–237
Table 8 Representative natural products with effect on GK activitya
Compound Activity a Control, activity Source Ref.
a Insulin: INS, Metformin: MET, Glibenclamide: GLB, Gliclazide: GLZ, Rosiglitazone: RSG, Commercial standard: CT, NM: not mentioned; a values are expressed as mean; b measured in diabetic rat hepatic tissue; c mmol of Pi liberated per h per protein; d units per g protein; e U per h per mg protein; f U mg−1 in the presence of g 20 and h 40 mg kg−1 of the compound; i μmol of glucose phosphorylated (U*) per h per mg protein; j μmol per min per mg protein; *activity in the presence of k 10, l 20, m 25, n 40, o 50, p 80, q 100, r 150, s 200, or t 300 mg kg−1 of the compound; u units per g protein; v nmol NAPDH per min per mg protein; w mU, mmol of substrate molecules converted by 1 mg protein per minute; x μM concentration of compound required to increase enzyme activity by 50%; y measured on isolated enzyme; z μg per mg of tissue.
Catechin
Catechin (6) 125.3b,c INS 3 IU kg−1; 118.2b,c Cassia fistula Linn (Fabaceae) 204
[thin space (1/6-em)]
Flavanones
Hesperidin (20) 149.8b,d GLB 100.0 mg kg−1; 153.0b,d Citrus spp. L. (Rutaceae) 208
Hesperetin (300) 0.4b,e GLB 1.0 mg kg−1; 0.4b,e 207
[thin space (1/6-em)]
Flavones
Amenthoflavone (301) 0.6b,f,*l; 1.1b,f,*n RSG 4.0 mg kg−1; 1.00b,f Selaginella tamariscina (P. Beauv.) Spring (Selaginellaceae) 202
Tangeretin-1 (302) 205.5b,i GLB 5.0 mg kg−1; 208.8b,i Poncirus trifoliata Raf. (Rutaceae) 210
[thin space (1/6-em)]
Isoflavones
Biochanin A (297) 0.3b,i GLB 600.0 μg kg; 0.4b,i Glycine max L. (Fabaceae) 203
[thin space (1/6-em)]
Flavanol
Kaempferol (56) ∼1.1b, j NM CT 209
[thin space (1/6-em)]
Terpenoids
D-Limonene (303) 0.2b,i,*o; 0.2b,i,*q; 0.2b,i,*q GLB 600.0 μg kg−1; 0.3b,i Citrus spp. L. (Rutaceae) 212
Geraniol (304) 138.6b,u GLZ 5.0 mg kg−1; 140.3b,u CT 213
Andrographolide (305) 0.3b,u MET 500.0 mg kg−1; 0.3b,u Andrographis paniculata (Burm.f.) Nees (Acanthaceae) 215
Rebaudioside A (306) 0.4b,i GLB 600.0 μg kg−1; 0.4b,i Stevia rebaudiana Bertoni (Asteraceae) 216
18β-Glycyrrhetinic acid (307) 0.2b,i,*l; 0.3b,i,*m; 0.2b,i,*o GLB 600.0 μg kg−1; 0.3b,i Glycyrrhiza glabra L. (Fabaceae) 218
Asiatic acid (308) ∼0.3b,i GLB 600.0 μg kg−1; ∼0.3b,i Centella asiatica L Urb. (Apiaceae) 219
Betulin (309) 0.3 and 0.6b,e MET 150.0 mg kg−1; 0.6b,e Striga orobanchioides Benth (Orobanchaceae) 220
Oleanolic acid (310) ∼7.0b,v INS 200.0 μg kg−1; ∼10b,v Syzygium aromaticum (L.) Merr. & L.M.Perry (Myrtaceae) 221
Meliacinolin (311) 2.9b,w GLB 4.0 mg kg−1; 3.1b,w Azadirachta indica A. Juss. (Meliaceae) 222
[thin space (1/6-em)]
Alkaloids
Berbamine (312) 200.1b,i MET 200.0 mg kg−1; 230.1b,i Berberis aristata DC. (Berberidaceae) 198
Betanin (313) 0.2b,i GLB 600.0 μg kg−1; 0.2b,i Beta vulgaris var. rubra L., sp. Pl., (Amaranthaceae) 199
Neotatarine (314) 5.9 μMx,y GKA22
2.1 μMx,y Acorus calamus L. (Acoraceae) 200
[thin space (1/6-em)]
Others
Esculetin (315) 0.2b,i,*k; 0.2b,i,*l; 0.3b,i,*n NM Matricaria chamomilla L. (Asteraceae) 205
Bellidifolin (316) 1.7b,*q,w; 1.8b,*s,w RSG 2.0 mg kg−1; 3.1b,w Swertia punicea Hemsl. (Gentianaceae) 224
Methylswertianin (317) 1.7b,*q,w; 1.8b,*s,w
Mangiferin (84) Increased activity y ∼25% above blank activity y Mangifera indica L. (Anacardiaceae) 225
α-Mangostin (318) 108.2b,*m,z; 122.0b,*o,z; 138.8b,*q,z GLB 10.0 mg kg−1; 135.4b,z Garcinia mangostana L. (Clusiaceae) 226
Protocatechuic acid (319) 0.3b,i; 0.4b,i; 0.3b,i GLB 600.0 μg kg−1; 0.4b,i Hibiscus sabdariffa L. (Malvaceae) 201
Fraxetin (320) 0.3b,i NM Fraxinus chinensis Roxb. (Oleaceae) 206
Tyrosol (321) 135.1b,i GLB 600.0 μg kg−1; 149.0b,i Olea europaea L. (Oleaceae) 211
3-Hydroxymethyl xylitol (322) 0.3b,i GLB 600.0 μg kg−1; 0.3b,i Casearia esculenta (Roxb.) (Salicaceae.) 223
Tatanan A (323) 1.9x,y GKA22 3.0 μMx,y Acorus tatarinowii Schott
(Araceae) 217
Tatanan B (324) 0.5x,y
Tatanan C (325) 0.2x,y
Trans-anethole (326) 135.6b,d GLB 600.0 μg kg; 3.1b,d Foeniculum vulgare Mill (Apiaceae) 214


Table 9 Representative natural products with effect on GK level expressiona
Compound Effect on GK expression Source Ref.
a CT: commercial standard; a evaluated in HepG2 cells; b evaluated in hepatic tissue; c evaluated in INS-1E cells; d GK mRNA abundance; e evaluated in pancreatic tissue.
Anthocyanins
Cyanidin 3-O-β-D-glucopyranoside (2) Increasea Morus spp. (Moraceae) 227
Cyanidin-3-O-α-D-rutinoside (327) Increasea
[thin space (1/6-em)]
Chalcone
Trilobatin (328) Increaseb Lithocarpus polystachyus Rehd. (Fagaceae) 231
[thin space (1/6-em)]
Flavanones
Naringenin (21) Increasec,d Citrus spp. L. (Rutaceae) 228
[thin space (1/6-em)]
Flavanol
Quercetin (68) Increasec,d CT 228
[thin space (1/6-em)]
Polyphenols
Caffeic acid (78) Increasec,d CT 228
Resveratrol (86) Reductiona Morus sp. L. (Moraceae) 229
Oxyresveratrol (88) Reductiona 227
Curcumin (329) Increasea Curcuma longa L. (Zingiberaceae) 229
Increasec 193
Phloridzin (330) Increaseb L. polystachyus Rehd. (Fagaceae) 231
Polydatin (331) Increaseb Polygonum cuspidatum Sieb. et Zucc (Polygonaceae) 233
[thin space (1/6-em)]
Triterpenoids
Ursolic acid (332) Increaseb Lycopersicon esculentum Mill. (Solaneacea) 230
Esculeoside A (333) Increaseb CT 234
[thin space (1/6-em)]
Alkaloids
Berberine (191) Increaseb Morus sp. L. (Moraceae) 227
[thin space (1/6-em)]
Others
1-Deoxynojrimycin (334) Increasea Morus sp. L. (Moraceae) 227
Phycocyanin (335) Increaseb,e Spirulina platensis Turpin ex Gomont (Spirulinaceae) 232


Among terpenoids, D-limonene (303) showed a significant hypoglycemic activity when given to STZ-diabetic rats (50–200 mg kg−1 day−1 per 45 days). GK activity was slightly increased compared to diabetic control.212 Geraniol (304) (200 mg kg−1 day−1 per 45 days), displayed moderated GK activation, since it increased the number of insulin-positive cells per islet in STZ-diabetic rats, thus significantly augmenting insulin levels.213 Interestingly, trans-anethole (326), a phenylpropanoid, also ameliorates the β-cells architecture in a similar model (80 mg kg−1 day−1 per 45 days), which increases glycogen level (44.47 ± 4.22 mg per g tissue respect to 22.73 ± 0.02 mg per g of diabetic control).214

The diterpenoid andrographolide (305) administered for 21 days (10 mg kg−1) did not affect insulin levels in STZ-diabetic rats; instead, it significantly increased glycogen levels with respect to diabetic animals (54.2 ± 4.7 mg per g tissue versus 33.4 ± 4.1 mg per g tissue), therefore suggesting extrapancreatic effects of GK activation.215 Rebaudioside A (306) showed potential GK activities, as its administration to STZ-diabetic rats (200 mg kg−1 day−1 per 45 days) protects pancreatic β-cells by decreasing oxidative stress, increasing insulin levels and glycogen content (45.01 ± 3.67 mg per g tissue versus 22.50 ± 1.82 mg per g tissue observed in diabetic control).216

There are two contradictory reports on the activity of sesquilignans tatanans A–C (323–325). Ni and co-workers (2011) indicated that these compounds were potent GK allosteric activators in vitro,217 while Xiao and co-workers (2013) refuted these findings.237 Thus, further investigations are required to clarify whether tatanans are GK activators, particularly under in vivo conditions.

Betulin (309),220 as well as 18β-glycyrrhetinic (307),218 asiatic (308),219 ursolic (332),234 and oleanolic (310)221 acids augmented GK activation and increased glycogen levels in diabetic rodents. Compounds 307 and 308 induce a significant augmentation of insulin levels, whilst triterpenoid 310 activated GK in a similar manner as insulin (used as positive control). Docking studies predicted that these triterpenoids, and the related β-amyrin, could be GK allosteric activators, but showed different interaction pattern than flavonoids.220 The predominant observed interactions were hydrophobic.235 The tetranortriterpenoid meliacinolin (311), daily administered for 28 days at a dose of 4 mg kg−1, increased both insulin and glycogen levels (17.86 ± 1.98 mg per g tissue and 3.34 ± 0.42 μU mL−1, respectively) when compared with STZ-diabetic controls (9.25 ± 1.65 mg per g tissue and 1.42 ± 0.18 μU mL−1, respectively) and in a similar fashion than glibenclamide, used as positive cotrol.222

GK activators modulates other metabolic pathways, as the amount of G6P is available for subsequent reactions. For example, in hepatocytes, G6P is dephosphorylated to glucose by glucose 6-phosphatase (G6Pase), a rate-limiting gluconeogenetic enzyme, which have an opposite effect than GK; thus GK and G6Pase have opposite actions and regulate each other activity.238 Kaempferol (56),209 berberine (191),192 amentoflavone (301),202 rebaudioside A (306),222 18β-glycyrrhetinic acid (307),238 meliacinolin (311),222 bellidifolin (316), methylswertianin (317),224 polydatin (331),233 and ursolic acid (332)234 activate GK and reduce both G6Pase activity and fasting glycemia.

In the liver, GK activators increase the activity of glycogen synthase (GS) and inhibit glycogen synthase kinase 3 (GSK-3β) promoting hepatic glycogen synthesis. There are reports of such activities for catechin (6),204 hesperidin (20),208 amentoflavone (301),202 tangeretin-1 (302),239 and tyrosol (321).211 Finally, G6P can be used in the last step of glycolysis controlled by pyruvate kinase (PK), an enzyme whose activity is reduced in diabetic rodent models. Several GK activators, such as amentoflavone (301),202 tangeretin-1 (302),239 asiatic acid (308),219 and betanin (313),199 activate PK.240

Since insulin controls hepatic GK expression, secondary metabolites able to active insulin pathways could increase GK levels. Thus, protein levels of different elements of the phosphatidylinositol-3-kinase (PI3K)/PKB canonical pathway can be monitored, including insulin receptor substrate-1 (IRS-1), PDK-1, Akt, and GSK-3β.241 Curcumin (329) increased phosphorylation of insulin receptor (IR), IRS-1, PI3K and Akt.193,222 Bellidifolin (316) and methylswertianin (317) increased the expression levels of IR-subunit α, IRS-1, and PI3K.224 Amentoflavone (301) also augmented the levels of phosphorylated Akt,202 and berberine (191) increased PI3K expression.192

Other studies on GK involved a series of in silico evaluations (molecular dynamics simulations and ADMET studies) of prenylated flavonoids, revealing their potential as GK activators.197

image file: d3np00007a-u36.tif

image file: d3np00007a-u37.tif

image file: d3np00007a-u38.tif

image file: d3np00007a-u39.tif

image file: d3np00007a-u40.tif

image file: d3np00007a-u41.tif

image file: d3np00007a-u42.tif

image file: d3np00007a-u43.tif

image file: d3np00007a-u44.tif

image file: d3np00007a-u45.tif

7. Fructokinase inhibitors

7.1. Structure of KHK and metabolic actions

Fructokinase (ketohexokinase, KHK, E.C. 2.7.1.3) is a 39 kDa dimeric enzyme belonging to the ribokinase-like superfamily with a high substrate specificity (Fig. 12). KHK exists in two isoforms: fructokinase A (KHK-A) and fructokinase C (KHK-C). Each monomer of KHK isoforms has two distinct domains: a central α/β-fold and a four-stranded β-sheet. The α/β-fold comprises a nine-stranded β-sheet flanked on each side by five α-helices. There is one active site per subunit in a cleft between the α/β-domain, and the four-stranded β-sheet that forms the dimer interface. AMP-PNP (adenylyl-imidodiphosphate) and fructose bind to opposite sites at the active-site cleft, being fructose-binding closer to the four-stranded β-sheet.242
image file: d3np00007a-f12.tif
Fig. 12 Ribbon representation of the human KHK complex with SO42− (https://www.rcsb.org/structure/2HQQ.

Although KHK-A is ubiquitous in most tissues, KHK-C is mainly expressed in the liver, kidney, and intestines and it is the main enzyme involved in fructose metabolism.243,244 KHK catalyzes the phosphorylation of fructose to fructose-1-phosphate (F1P) using ATP and a potassium ion (K+) (Fig. 13). KHK-C has a higher affinity for fructose phosphorylation than KHK-A.245,246 Unlike GK, KHK catalytic actions are not controlled by insulin but by fructose.247


image file: d3np00007a-f13.tif
Fig. 13 Reaction mechanism of glucose phosphorylation catalyzed by KHK.

Fructose enters rapidly into the liver using the glucose transporter-2 (GLUT-2) and is converted to F1P by KHK. F1P is a substrate of aldolase B, resulting in phosphotrioses used to synthesized glucose, lactate, and fatty acids. Increased fructose intake mainly due to the uncontrolled ingestion of sweetened beverages and highly processed foods, leads to an augmentation in triglyceride synthesis, as glycolysis intermediates are accumulated and then to glycerol-3-phosphate.247 Due to fructose phosphorylation, KHK triggers the depletion of ATP and facilitates uric acid liver and fat accumulation.248 Since KHK is not inhibited by its catalytic product nor regulated allosterically, there is a need to search for KHK inhibitors, as an alternative to diminishing fatty acids and triglyceride accumulation.249

7.2. Assay methods

The activity of KHK can be assessed in vitro using several assays. Lee and co-workers249 reported a 3-step cell-free enzymatic reactions which monitors (i) ADP formation related to fructose phosphorylation, (ii) pyruvate increase as a result of ADP and P-enol pyruvate reaction, and finally (iii) measures the stoichiometric decrease in NADH absorbance at 340 nm.249 Maryanoff and coworkers reported a method using HepG2 cells, which, after being exposed to test compounds, received fructose and were further processed to obtain cell lysates that were used to quantify F1P levels by means of liquid chromatography-mass spectrometry.250,251 Another method to assess KHK activity is to determine fructose-induced ATP depletion. This assay is conducted in cell lysates of HepG2 cells overexpressing KHK-C. ATP level, measured using an ATP assay kit, and ATP depletion is proportional to KHK activity.249

7.3. KHK inhibitors from medicinal plants

In 2016, Le and coworkers published reported 406 plant extracts and 1200 purified phytochemicals activity against KHK-C. Promising result were obtained with Angelica archangelica (Apiaceae), Scutellaria baicalensis (Lamiaceae), Petroselinum crispum (Apiaceae), and Garcinia mangostana (Clusiaceae). Fourteen secondary metabolites derived from these extracts were also analyzed (Table 10).249
Table 10 Representative natural products with FK inhibitory activity
Compound IC50 (μM) Control, inhibition (%) Source Ref.
Chalcones
Methoxy isobavachalcone (336) 0.2 4-(Hydroxymercuri) benzoic acid sodium salt, 100% at 0.2 mM Psoralea corylifolia (L.) Medik. (Fabaceae) 249 and 250
Isobavachalcone (337) 5.7
[thin space (1/6-em)]
Flavone
Oroxylin A (338) 8.2 Andrographis paniculate Burm. F. (Acanthaceae)
Apigenin 7-glucuronide (339) 16.1 Scutellaria baicalensis Georgi (Lamiaceae)
Apiin (340) 21.5 Petroselinum crispum (Mill.) Fuss (Apiaceae)
Mulberrin (341) 31.2 Morus alba L. (Moraceae)
[thin space (1/6-em)]
Other polyphenols
Cratoxycarborenone E (342) 1.0 Cratoxylum prunifolium (Wall.) Dyer (Hypericaceae)
α-Mangostin (318) and γ-mangostin (343) mixture 1.5
Osthenol (344) 7.8 Angelica archangelica L. (Apiaceae)
Osthole (345) 0.7
5,7-Dimethoxy-8-prenylcoumarin (346) 10.3 Diospyros attenuate (Thw.) (Ebaneaceae)
Flavaspidic acid AB (347) 29.5 Pteris wallichiana C. Agardh (Pteridaceae)
Swietenocoumarin B (348) 58.4 Chloroxylon swietenia DC. (Rutaceae)


The activity of the compounds ranged from 16.1 to 93.4% of inhibition. Among the prenylated chalcones 336 and 337, the methoxy isobavachalcone (336) was the most potent (IC50 = 0.2 μM related to depletion of hepatic ATP).249 This metabolite also inhibits adipogenesis and contribute to triglycerides homeostasis, acting as a non-competitive inhibitor of acyl-coenzyme A: cholesterol acyltransferase (ACAT), and as an inhibitor of diacylglycerol acyltransferase (DGAT).252 Four flavones (338–341) have been reported to be FK inhibitors, oroxylin A (338) is the most potent (42.1% inhibition, IC50 = 8.2 μM). Other polyphenols such as cratoxyarborenone E (342) (61.7% inhibition, IC50 = 1.0 μM), and a mixture of α-/γ-mangostin (318/343, 90.4% inhibition, IC50 = 1.5 μM) showed prominent inhibitory activities against KHK.249 Prenylated coumarins such as osthenol (344), osthole (345) and the 2-methoxylated derivative (346) also displayed significant KHK inhibition, being 345 twice as potent as 344. Osthole (345) is a hypoglycemic molecule that acts as a dual PPARα/γ and AMPK activator, thus inducing the activities of acyl-CoA synthetases and acetyl CoA carboxylase, respectively, without lowering blood insulin or lipid levels.253,254 Along these lines, prenylated xanthones α-mangostin (318) and γ-mangostin (343) evaluated as a mixture, inhibited almost all KHK activity. Investigations using STZ-diabetic animals suggest that 318 is a hypoglycemic agent that induces insulin secretion, increased GK activity, reduces fructose-1-6-biphosphatase and glucose-6-phosphatase activities,226,255 and increases leptin and glucose transporter-4 (GLUT-4) expressions. γ-Mangostin has also shown to increase glucose uptake and diminishes saccharide digestion inhibiting intestinal α-amylase and α-glucosidase enzymes.256 A common structural feature among the most active compounds is the presence of a prenylated side chain (either an isoprenyl, a geranyl, or a 1,1-dimethylallyl moiety). Therefore, further studies on natural products should be conducted to assess binding interactions with this enzyme using computational or crystallographic tools.257

image file: d3np00007a-u46.tif

8. Concluding remarks and future directions

This review has summarized the natural compounds reported in the literature from 2000 to 2022 having in vitro or in vivo DPP-IV, DGAT, FBPase, GK, or KHK activities (Tables 1–10). No therapeutic agents affecting DGAT, FBPase, GK, or KHK have been developed yet.

The high number of reported flavonoids (Fig. 14) as inhibitors or activators of such enzymes and their non-selective actions is not surprising because it is well-documented that flavonoids are polypharmacological (polyvalent) agents; this is, they can interact with two or more molecular targets simultaneously, thus affecting multiple biological processes.258 Therefore modulating several mechanisms involved in glucose homeostasis, acting through different enzymes.258 Pharmacological polyvalence can arise from conserved binding sites in different proteins recognized by different compound scaffolds or from the compound's structural motifs (auxophoric moieties) prone to trigger multiple ligand-target interactions.259 In this context, flavonoids are listed as pan-assay interference compounds (PAINS) because most of them are phenolic. There are multiple reports where PAINS have shown to give false positives in assays, thus misleading investigations and generating several overstatements on observed activities. Therefore, a detailed structure–activity relationship analysis is necessary to identify such compounds' pharmacophoric and auxophoric moieties, thus leading to rational drug design. PAINS presence in drugs is common, as shown by the fact that almost 5% of FDA-approved drugs contain PAINS-recognized scaffolds, comprising both natural products and synthetic drugs. Concerns regarding PAINS in natural products must be overcome or confirmed using multiple validated and complementary experimental tools, such as in vitro, in silico, and in vivo testing.259–261


image file: d3np00007a-f14.tif
Fig. 14 Different natural product types target key enzymes in glucose metabolism.

For instance, we found significant differences in some compounds' inhibitory activity against DPP-IV during this revision. These discrepancies might be attributed to variations in the methodology, enzymes, and conditions to run the tests. Furthermore, different authors' docking analyses of many natural products evaluated in vitro, particularly flavonoids, yielded contradictory results (Table 1). This situation precluded any conclusion about the structure–activity relationship among the same type of compounds. Perhaps the next generation of DPP-IV inhibitors from natural sources could emerge from foods, as many articles emphasize the relevance of the consumption of dietary proteins from soybean, rice, amaranth, draft beer, broccoli, Brassica spp, macroalgae, yam species, and beans, which upon digestion, yield inhibitory DPP-IV peptides (Fig. 14).

Those compounds listed in Tables 5 and 6 targeting DGAT showed inhibitory activities from 0.46 to >250 μM. The lack of therapeutic drugs aiming DGAT made that the positive control used in most cases was kuridin (200), a natural chalcone from Sophora flavescens, which along with aphadilactone C (244), glabrol (203), kurarinone (204), kurarinol (205), amidepsine A (270), and phenylpyropenes C (280) are the most active inhibitory natural products against DGAT. Aphadilactone C (244) warrants further investigation as a DGAT-1 inhibitor due to its high selectivity, potency, and unusual scaffold.

Regarding the FBPase inhibitors, all compounds reviewed were flavonoids, but usnic acid (299), a well-known lichen metabolite. The most active were baicalein (27), scutellarein (300), herbacetin (308), and gossypetin (303), whose activities were comparable with the positive control, AMP (IC50 of 2.6 μM). There is an urgent need for a more simple and robust in vitro analysis for detecting active compounds.

GK activators (Tables 8 and 9) could bind to the allosteric site or modulate protein expression. Flavonoids amentoflavone (301), kaempferol (56), and quercetin (68) are known allosteric activators of GK. In contrast, triterpenoids 307–318 could activate GK by an allosteric mechanism different from that of flavonoids. It is relevant to mention that secretagogue natural products that interfere with the insulin pathway could modify GK activity, as documented for compounds 6, 56, 29, 191, 297, 301, 302, 316, and 317. Finally, natural KHK inhibitors, such as 318, 336, 343, 344, and 354, showed in vivo hypoglycemic activity and regulated triglyceride synthesis.

Interestingly, multi-target activities displayed by natural products are an asset in developing drugs to treat complex and multi-factorial diseases such as TIIDM.262 This review shows that natural products could improve glucose homeostasis by modulating related proteins and pathways. Therefore, a single chemical identity could effectively improve insulin resistance and hyperglycemia due to different etiologies. In the context of the worldwide spread of diabetic complications, an intensive search for new lead compounds for developing novel pharmacological therapeutics is extremely important, whereas clinical trials are still lacking. In this regard, it is important to remember that calculated IC50 using in vitro models assesses ligand binding affinities to enzymes rather than ligand-target kinetics or ligand-target removal. Therefore, a straightforward correlation between preclinical (in vitro or in vivo) and clinical potency is not to be expected.263

For instance, in vivo level of receptor occupancy required to achieve maximal efficacy is related to the target's mechanism of action and its downstream signaling components rather than the quantity of drug needed for activation. Thus, compounds activating multiple elements in a signaling pathway have better chances to induce the desired pharmacological effect by acting additively or synergically, as proven for natural compounds. However, in vivo pharmacokinetics significantly impact the observed potency, as most of the natural compounds herein described possess low bioavailability. Thus, effective clinical concentrations will be hard to achieve without making structural modifications that could improve their absorption, reduce their protein-binding or modify their elimination, well-known optimizations conducted to obtain drugs from bioactive molecules.

Further studies are necessary to fully understand the mode of action of natural products targeting multiple enzymes, establish structure–activity relationships, therapeutical concentrations, and required scaffold modifications to improve pharmacokinetics or potency.

The continuous research of plants used in traditional and complementary medicines and other natural sources might lead to the discovery of new successful antidiabetic drugs, as before with metformin, acarbose, exenatide, and gliptins, all based on natural products, all belonging to different categories of natural products. As shown in Fig. 14, alkaloids and terpenoids could be good candidates for developing modulators of DPP-IV, DGAT, FBPase, GK, or KHK.

9. Conflicts of interest

There are no conflicts to declare.

10. Acknowledgements

This work was supported by grants of Dirección General de Asuntos de Personal Académico (DGAPA, IA205621 and IN203523), Consejo Nacional de Ciencia y Tecnología (CONACyT, CY011226), and L’Oréal-UNESCO-AMC-CONALMEX awarded to R.M.

11. References

  1. International Diabetes Federation. IDF Diabetes Atlas, International Federation of Diabetes, Brussels, Belgium, 10th edn, 2021, ISBN: 978-2-930229-98-0 Search PubMed .
  2. American Diabetes Association Professional Practice Committee, Diabetes Care, 2022, 45, S125–S143 CrossRef PubMed .
  3. A. D. Dahlén, G. Dashi, I. Maslov, M. M. Attwood, J. Jonsson, V. Trukhan and H. B. Schiöth, Front. Pharmacol., 2022, 12, 807548 CrossRef PubMed .
  4. C. J. Bailey, Diabetologia, 2017, 60, 1566–1576 CrossRef CAS PubMed .
  5. E. C. Chao and R. R. Henry, Nat. Rev. Drug Discovery, 2010, 9, 551–559 CrossRef CAS PubMed .
  6. A. E. Martin and P. A. Montgomery, Am. J. Health-Syst. Pharm., 1996, 53, 2277–2290 CrossRef CAS PubMed .
  7. A. Mehanna, Future Med. Chem., 2013, 5, 411–430 CrossRef CAS PubMed .
  8. S. Sugimoto, H. Nakajima, K. Kosaka and H. Hosoi, Nutr. Metab., 2015, 12, 51 CrossRef PubMed .
  9. D. J. Newman and G. M. Cragg, J. Nat. Prod., 2012, 75, 311–335 CrossRef CAS PubMed .
  10. A. C. Rufer, Drug Discovery, 2021, 26, 875–886 CAS .
  11. R. Thoma, B. Loffler, M. Stihle, W. Huber, A. Ruf and M. Hennig, Structure, 2003, 11, 947–959 CrossRef CAS PubMed .
  12. C. A. Abbott, G. W. McCaughan and M. D Gorrell, FEBS Lett., 1999, 458, 278–284 CrossRef CAS PubMed .
  13. S. Arulmozhiraja, N. Matsuo, E. Ishitsubo, S. Okazaki, H. Shimano and H. Tokiwa, PLoS One, 2016, 11, e0166275 CrossRef PubMed .
  14. H. B. Rasmussen, S. Branner, F. C. Wiberg and N. Wagtmann, Nat. Struct. Biol., 2003, 10, 19–25 CrossRef CAS PubMed .
  15. R. Ebert and W. Creutzfeldt, Diabetes/Metab. Res. Rev., 1987, 3, 1–26 CrossRef CAS PubMed .
  16. L. Zhu, C. Tamvakopoulos, D. Xie, J. Dragovic, X. Shen, J. E. Fenyk-Melody, K. Schmidt, A. Bagchi, P. R. Griffin, N. A. Thornberry and R. S. Roy, J. Biol. Chem., 2003, 278, 22418–22423 CrossRef CAS PubMed .
  17. J. J. Holst, Physiol. Rev., 2007, 87, 1409–1439 CrossRef CAS PubMed .
  18. D. Röhrborn, N. Wronkowitz and J. Eckel, Front. Immunol., 2015, 6, 386–406 Search PubMed .
  19. C. F. Deacon, Front. Endocrinol., 2019, 10, 80 CrossRef PubMed .
  20. R. Eissele, R. Goke, S. Willemer, H. P. Harthus, H. Vermeer, R. Arnold and B. Goke, Eur. J. Clin. Invest., 1992, 22, 283–291 CrossRef CAS PubMed .
  21. T. Nagatsu, M. Hino, H. Fuyamada, T. Hayakawa and S. Sakakibara, Anal. Biochem., 1976, 74, 466–476 CrossRef CAS PubMed .
  22. E. Boonacker, S. Elferink, A. Bardai, B. Fleischer and C. J. Van Noorden, J. Histochem. Cytochem., 2003, 51, 959 CrossRef CAS PubMed .
  23. X. L. Wu, Y. Wang and X. P. Zhao, Zhongguo Zhongyao Zazhi, 2016, 41, 1241–1245 Search PubMed .
  24. J. Zhang, Y. Liu, J. Lv, Y. Cao and G. Li, Microchim. Acta, 2015, 182, 281–288 CrossRef CAS .
  25. N. Xia, X. Wang, X. Wang and B. Zhou, Materials, 2016, 9, 857 CrossRef PubMed .
  26. A. D. Tinoco, D. M. Tagore and A. Saghatelian, J. Am. Chem. Soc., 2010, 132, 3819–3830 CrossRef CAS PubMed .
  27. J. Liu, X. Cheng and L. Fu, Anal. Methods, 2012, 4, 1797–1805 RSC .
  28. L. Gu, T. Tian, L. Xia, G. Chou and Z. Wang, J. Planar Chromatogr.–Mod. TLC, 2019, 32, 447–451 CrossRef CAS .
  29. L. Guasch, M. J. Ojeda, N. González-Abuín, E. Sala and A. Cereto-Massague, PLoS One, 2012, 7, e44971 CrossRef CAS PubMed .
  30. D. Istrate and L. Crisan, Symmetry, 2022, 14, 1842 CrossRef CAS .
  31. J.-L. Ríos, I. Andújar, G. R. Schinella and F. Francini, Planta Med., 2019, 85, 825–839 CrossRef PubMed .
  32. R. K. Singla, R. Kumar, S. Khan, Mohit, K. Kumari and A. Garg, Curr. Protein Pept. Sci., 2019, 20, 1218–1225 CrossRef CAS PubMed .
  33. F. Les, G. Cásedas, C. Gómez, C. Moliner, M. S. Valero and V. López, J. Physiol. Biochem., 2020, 77, 109–131 CrossRef PubMed .
  34. M. Ajebli, H. Khan and M. Eddouks, Endocr., Metab. Immune Disord.: Drug Targets, 2021, 21, 111–130 CrossRef CAS PubMed .
  35. S. Shaikh, E.-J. Lee, K. Ahmad, S.-S. Ahmad, J.-H. Lim and I. Choi, Pharmaceuticals, 2021, 14, 591 CrossRef CAS PubMed .
  36. A.-K. Singh, D. Yadav, N. Sharma and J.-O. Jin, Pharmaceuticals, 2021, 14, 586 CrossRef CAS PubMed .
  37. S. Chhabria, S. Mathur, S. Vadakan, D. K. Sahoo, P. Mishra and B. Paital, Front. Endocrinol., 2022, 13, 1027237 CrossRef PubMed .
  38. Z. Li, J. Tian, Z. Cheng, W. Teng, W. Zhang, Y. Bao, Y. Wang, B. Song, Y. Chen and B. Li, Crit. Rev. Food Sci. Nutr., 2022, 25, 1–18 Search PubMed .
  39. C. Proença, D. Ribeiro, M. Freitas, F. Carvalho and E. Fernandes, Crit. Rev. Food Sci. Nutr., 2022, 62, 4095–4151 CrossRef PubMed .
  40. H. Sajal, S. M. Patil, R. Raj, A. M. Shbeer, M. Ageel and R. Ramu, Molecules, 2022, 27, 5133 CrossRef CAS PubMed .
  41. S. Supandi, M. S. Wulandari, E. Samsul, A. Azminah, R. Y. Purwoko, H. Herman, H. Kuncoro, A. Ibrahim, N. S. S. Ambarwati, R. Rosmalena, R. N. Azizah, S. Paramita and I. Ahmad, J. Adv. Pharm. Technol. Res., 2022, 13, 207–215 CAS .
  42. J. Fan, M. H. Johnson, M. A. Lila, G. Yousef and E. Gonzalez de Mejia, J. Evidence-Based Complementary Altern. Med., 2013, 2013, 479505 Search PubMed .
  43. F. Gao, Y. Fu, J. Yi, A. Gao, Y. Jia and S. Cai, J. Agric. Food Chem., 2020, 68, 12141–12151 CrossRef CAS PubMed .
  44. J. Pan, Q. Zhang, C. Zhang, W. Yang, H. Liu, Z. Lv, J. Liu and Z. Jiao, Front. Nutr., 2022, 9, 892426 CrossRef PubMed .
  45. T. Morikawa, K. Ninomiya, J. Akaki, N. Kakihara, H. Kuramoto, Y. Matsumoto, T. Hayakawa, O. Muraoka, L.-B. Wang, L.-J. Wu, S. Nakamura, M. Yoshikawa and H. Matsuda, J. Nat. Med., 2015, 69, 494–506 CrossRef CAS PubMed .
  46. B.-R. Kim, S. B. Paudel, J.-W. Nam, C. H. Jin, I.-S. Lee and A.-R. Han, Molecules, 2020, 25, 4370 CrossRef CAS PubMed .
  47. M. N. Sarian, Q. U. Ahmed, S. Z. Mat So’ad, A. M. Alhassan, S. Murugesu, V. Perumal, S. N. A. S. Mohamad, A. Khatib and J. Latip, BioMed Res. Int., 2017, 2017, 8386065 Search PubMed .
  48. Y. Fujimura, M. Watanabe, T. Morikawa-Ichinose, K. Fujino, M. Yamamoto, S. Nishioka, C. Inoue, F. Ogawa, M. Yonekura, A. Nakasone, M. Kumazoe and H. Tachibana, J. Agric. Food Chem., 2022, 70, 6455–6466 CrossRef CAS PubMed .
  49. V. C. S. R. Chittepu, P. Kalhotra, T. Osorio-Gallardo, C. Jiménez-Martínez, R. R. Robles-de la Torre, T. Gallardo-Velazquez and G. Osorio-Revilla, Molecules, 2019, 24, 3887 CrossRef CAS PubMed .
  50. A. M. Bower, L. M. Real Hernandez, M. A. Berhow and E. Gonzalez de Mejia, J. Agric. Food Chem., 2014, 62, 6147–6158 CrossRef CAS PubMed .
  51. H. Yaribeygi, S. L. Atkin and A. Sahebkar, J. Cell. Biochem., 2019, 120, 10909–10913 CrossRef CAS PubMed .
  52. M. F. Khalid, K. Rehman, K. Irshad, T. A. Chohan and M. S. H. Akash, Dose-Response, 2022, 20, 15593258221093275 CrossRef CAS PubMed .
  53. Q. Huang, J.-J. Chen, Y. Pan, X.-F. He, Y. Wang, X.-M. Zhang and C.-A. Geng, J. Pharm. Biomed. Anal., 2021, 198, 113998 CrossRef CAS PubMed .
  54. B.-R. Kim, H. Y. Kim, I. Choi, J.-B. Kim, C. H. Jin and A.-R. Han, Molecules, 2018, 23, 1998 CrossRef PubMed .
  55. B. Hou, M.-T. Kuang, X.-Q. Chi, J.-Y. Li, L. Yang, Z.-H. Liu, M.-Y. Yuan, Q.-H. Kong, J.-M. Hu and J. Zhou, ChemistrySelect, 2018, 3, 10864–10868 CrossRef CAS .
  56. B. T. Zhao, D. D. Le, P. H. Nguyen, M. Y. Ali, J.-S. Choi, B. S. Min, H. M. Shin, H. I. Rhee and M. H. Woo, Chem.-Biol. Interact., 2016, 253, 27–37 CrossRef CAS PubMed .
  57. P. Kalhotra, V. C. S. R. Chittepu, G. Osorio-Revilla and T. Gallardo-Velázquez, Int. J. Mol. Sci., 2019, 20, 1228 CrossRef CAS PubMed .
  58. T. Eidenberger, M. Selg and K. Krennhuber, Fitoterapia, 2013, 89, 74–79 CrossRef CAS PubMed .
  59. M. Li, X. Bao, X. Zhang, H. Ren, S. Cai, X. Hu and J. Yi, LWT–Food Sci. Technol., 2022, 162, 113467 CrossRef CAS .
  60. R. Praparatana, P. Maliyam, L. R. Barrows and P. Puttarak, Molecules, 2022, 27, 2393 CrossRef CAS PubMed .
  61. A.-K. Singh, P. K. Patel, K. Choudhary, J. Joshi, D. Yadav and J.-O. Jin, Biomolecules, 2020, 10, 207 CrossRef CAS PubMed .
  62. Y. Fu, X. Liu, Q. Ma, J. Yi and S. Cai, Int. J. Food Sci., 2022, 57, 2656–2669 CrossRef CAS .
  63. B.-R. Kim, P. Thapa, H. M. Kim, C. H. Jin, S. H. Kim, J.-B. Kim, H. Choi, A.-R. Han and J.-W. Nam, ACS Omega, 2020, 5, 4050–4057 CrossRef CAS PubMed .
  64. N. S. S. Chalichem, S. Jupudi, V. R. Yasam and D. Basavan, J. Ayurveda Integr. Med., 2021, 12, 663e672 Search PubMed .
  65. T. Tuersuntuoheti, F. Pan, M. Zhang, Z. Wang, J. Han, Z. Sun and W. Song, J. Food Process. Preserv., 2022, 46, e16808 CAS .
  66. G. Cásedas, F. Les, M. P. Gómez-Serranillos, C. Smithc and V. López, Food Funct., 2017, 8, 4187–4193 RSC .
  67. F. O. Balogun, K. Naidoo, J. O. Aribisala, C. Pillay and S. Sabiu, Metabolites, 2022, 12, 937 CrossRef CAS PubMed .
  68. Z. Wang, L. Yang, H. Fan, P. Wu, F. Zhang, C. Zhang, W. Liu and M. Li, PeerJ, 2017, 5, e3283 CrossRef PubMed .
  69. A. Quek, N. K. Kassim, P. C. Lim, D. C. Tan, M. A. M. Latif, A. Ismail, K. Shaari and K. Awang, Pharm. Biol., 2021, 59, 964–973 CrossRef CAS PubMed .
  70. S. Bo, S. K. Chang, Y. Shan, Y. Chen, H. Liu, B. Li, Y. Jiang, H. Zhu and B. Yang, Food Res. Int., 2022, 157, 111275 CrossRef CAS PubMed .
  71. W. Nuankaew, A. Heemman, C. Wattanapiromsakul, J. H. Shim, N. W. Kim, T. Yasmin, S. Y. Jeong, Y. H. Nam, B. N. Hong, S. Dej-Adisai and T. H. Kang, J. Nat. Med., 2021, 75, 520–531 CrossRef CAS PubMed .
  72. C. Proença, M. Freitas, D. Ribeiro, S. M. Tomé, A. N. Araújo, A. M. S. Silva, P. A. Fernandes and E. Fernandes, Food Funct., 2019, 10, 5718–5731 RSC .
  73. M. Á. Martín and S. Ramos, Cells, 2021, 10, 1474 CrossRef PubMed .
  74. A. K. Jugran, S. Rawat, H. P. Devkota, I. D. Bhatt and R. S. Rawal, Phytother. Res., 2021, 35, 223–245 CrossRef PubMed .
  75. M. Naveed, V. Hejazi, M. Abbas, A. A. Kamboh, G. J. Khan, M. Shumzaid, F. Ahmad, D. Babazadeh, X. FangFang, F. Modarresi-Ghazani, L. WenHua and Z. XiaoHui, Biomed. Pharmacother., 2018, 97, 67–74 CrossRef CAS PubMed .
  76. M. Martorell, N. Castro, M. Victoriano, X. Capó, S. Tejada, S. Vitalini, R. Pezzani and A. Sureda, J. Evidence-Based Complementary Altern. Med., 2021, 20, 3313419 Search PubMed .
  77. Y. L. Ngo, C. H. Lau and L. S. Chua, Food Chem. Toxicol., 2018, 121, 687–700 CrossRef CAS PubMed .
  78. A. B. Nongonierma and R. J. FitzGerald, Peptides, 2013, 39, 157–163 CrossRef CAS PubMed .
  79. T.-Y. Wang, C.-H. Hsieh, C.-C. Hung, C.-L. Jao, M.-C. Chen and K.-C. Hsu, J. Funct. Foods, 2015, 19, 330–340 CrossRef CAS .
  80. A. Zambrowicz, E. Eckert, M. Pokora, Ł. Bobak, A. Dabrowska, M. Szołtysik, T. Trziszka and J. Chrzanowska, RSC Adv., 2015, 5, 10460–10467 RSC .
  81. R. Liu, J. Cheng and H. Wu, Int. J. Mol. Sci., 2019, 20, 463 CrossRef CAS PubMed .
  82. T. Wenhui, H. Shumin, Z. Yongliang, S. Liping and Y. Hua, J. Sci. Food Agric., 2022, 102, 1085–1094 CrossRef PubMed .
  83. T. Zhou, Z. Liu, J. Pei, D. Pan, X. Gao, Y. Dang and Y. Zhao, J. Sci. Food Agric., 2021, 69, 10885–10892 CAS .
  84. F. Xu, E. G. D. Mejia, H. Chen, K. Rebecca, M. Pan, R. He, Y. Yao, L. Wang and X. Ju, J. Food Biochem., 2020, 44, e13406 CAS .
  85. A. Iwaniak, M. Darewicz and M. Minkiewicz, Compr. Rev. Food Sci. Food Saf., 2018, 17, 63–81 CrossRef CAS PubMed .
  86. T. Hatanaka, Y. Inoue, J. Arima, Y. Kumagai, H. Usuki, K. Kawakami, M. Kimura and T. Mukaihara, Food Chem., 2012, 134, 797–802 CrossRef CAS PubMed .
  87. L. Mojica, D. A. Luna-Vital and E. González de Mejía, J. Sci. Food Agric., 2017, 97, 2401–2410 CrossRef CAS PubMed .
  88. P. A. Harnedy, M. B. O'Keeffe and R. J. Fitzgerald, Food Chem., 2015, 172, 400–406 CrossRef CAS PubMed .
  89. C. Lammi, C. Bollati, S. Ferruzza, G. Ranaldi, Y. Sambuy and A. Arnoldi, Nutrients, 2018, 10, 1082 CrossRef PubMed .
  90. Y.-S. Lin, C.-H. Han, S.-Y. Lin and W.-C. Hou, J. Agric. Food Chem., 2016, 64, 6451–6458 CrossRef CAS PubMed .
  91. A. B. Nongonierma and R. J. FitzGerald, J. Food Biochem., 2019, 43, e12451 CrossRef PubMed .
  92. H. Umezawa, T. Aoyagi, K. Ogawa, H. Naganawa, M. Hamada and T. Takeuchi, J. Antibiot., 1984, 37, 422–425 CrossRef CAS PubMed .
  93. H. Hiramatsu, A. Yamamoto, K. Kyono, Y. Higashiyama, C. Fukushima, H. Shima, S. Sugiyama, K. Inaka and R. Shimizu, Biol. Chem., 2004, 385, 561–564 CAS .
  94. J. Rahfeld, M. Schierhorn, B. Hartrodt, K. Neubert and J. Heins, Biochim. Biophys. Acta, 1991, 1076, 314–316 CrossRef CAS PubMed .
  95. X. Ye, L. Xiong, Q. Fu, B. Wang, Y. Wang, K. Zhang, J. Yang, F. Kantawong, W. Kumsaiyai, J. Zhou, C. Lan, J. Wu and J. Zeng, J. Ethnopharmacol., 2022, 292, 115203 CrossRef CAS PubMed .
  96. A. J. Velarde-Salcedo, A. Barrera-Pacheco, S. Lara-González, G. M. Montero-Morán, A. Díaz-Gois, E. González de Mejía and A. P. Barba de la Rosa, Food Chem., 2013, 136, 758–764 CrossRef CAS PubMed .
  97. L. Mojica, D. A. Luna-Vitala and E. González de Mejía, J. Sci. Food Agric., 2017, 97, 2401–2410 CrossRef CAS PubMed .
  98. S. Bleakley, M. Hayes, N. O’ Shea, E. Gallagher and T. Lafarga, Foods, 2017, 6, 108 CrossRef PubMed .
  99. A. B. Nongonierma, C. Mazzocchi, S. Paolella and R. J. FitzGerald, Food Res. Int., 2017, 94, 79–89 CrossRef CAS PubMed .
  100. Q. Tao, K. Ma, Y. Yang, K. Y. Wang, B. Chen, Y. Huang, J. Han, L. Bao, X.-B. Liu, Z. Yang, W.-B. Yin and H. Liu, J. Org. Chem., 2016, 81, 9867–9877 CrossRef CAS PubMed .
  101. K. R. R. Rengasamy, L. P. Slavětínská, M. G. Kulkarni, W. A. Stirk and J. Van Staden, Algal Res., 2017,(25), 178–183 CrossRef .
  102. S.-M. Zhao, G.-X. Chou, Q.-S. Yang, W. Wang and J.-L. Zhou, Org. Biomol. Chem., 2016, 14, 3510–3520 RSC .
  103. A. Khan, I. Khan, S. A. Halim, N. U. Rehman, N. Karim, W. Ahmad, M. Khan, M. Csuk and A. Al-Harrasi, Biomed. Pharmacother., 2022, 147, 112669 CrossRef CAS PubMed .
  104. P. S. Suresh, P. P. Singh, Y. S. Padwad and U. Sharma, J. Pharm. Pharmacol., 2021, 73, 487–495 CrossRef PubMed .
  105. S. Saleem, L. Jafri, I. U. Haq, L. C. Chang, D. Calderwood, B. D. Green and B. Mirza, J. Ethnopharmacol., 2014, 156, 26–32 CrossRef CAS PubMed .
  106. E. Kato, K. Kawakami and J. Kawabata, J. Enzyme Inhib. Med. Chem., 2018, 33, 106–109 CrossRef CAS PubMed .
  107. C. Lammi, M. Bartolomei, C. Bollati, L. Cecchi, M. Bellumori, E. Sabato, G. Vistoli, N. Mulinacci and A. Arnoldi, Antioxidants, 2021, 10, 1133 CrossRef CAS PubMed .
  108. L. Gu, T. Tian, L. Xia, G. Chou and Z. Wang, J. Planar Chromatogr.–Mod. TLC, 2019, 32, 447–451 CrossRef CAS .
  109. K. Budipramana, J. Junaidin, K. R. Wirasutisna, Y. B. Pramana and S. Sukrasno, Sci. Pharmacol., 2019, 87, 21 CrossRef CAS .
  110. M. J. Ojeda-Montes, A. Ardid-Ruiz, S. Tomás-Hernández, A. Gimeno, A. Cereto-Massagué, R. Beltrán-Debón, M. Mulero, S. Garcia-Vallvé, G. Pujadas and C. Valls, Future Med. Chem., 2017, 9, 2129–2146 CrossRef CAS PubMed .
  111. S. Naik, N. Deora, S. K. Pal, M. Z. Ahmed, A. S. Alqahtani, P. K. Shukla, K. Venkatraman and S. Kumar, J. Mol. Recognit., 2022, 35, e2983 CrossRef CAS PubMed .
  112. B. Adhikari, J. Chem., 2021, 2021, 2691525 Search PubMed .
  113. S. Krishnan, K. Chakraborty and M. Joy, J. Food Biochem., 2019, 43, e12824 CrossRef PubMed .
  114. F. Makkar and K. Chakraborty, Med. Chem. Res., 2018, 27, 1245–1259 CrossRef CAS .
  115. R. Costante, A. Stefanucci, S. Carradori, E. Novellino and A. Mollica, Expert Opin. Ther. Pat., 2015, 25, 209–236 CrossRef CAS PubMed .
  116. H. Hussain, G. Abbas, I. R. Green and I. Ali, Expert Opin. Ther. Pat., 2019, 29, 535–553 CrossRef CAS PubMed .
  117. R. Naik, B. W. Obiang-Obounou, M. Kim, Y. Choi, H. S. Lee and K. Lee, ChemMedChem, 2014, 9, 2410–2424 CrossRef CAS PubMed .
  118. X. Sui, K. Wang, N. L. Gluchowski, S. D. Elliott, M. Liao, T. C. Walther and R. V. Farese Jr, Nature, 2020, 581, 323–328 CrossRef CAS PubMed .
  119. L. Wang, H. Qian, Y. Nian, Y. Han, Z. Ren, H. Zhang, L. Hu, B. V. V. Prasad, A. Laganowsky, N. Yan and M. Zhou, Nature, 2020, 581, 323–328 CrossRef PubMed .
  120. A. M. Birch, S. Birtles, L. K. Buckett, P. D. Kemmitt, G. J. Smith, T. J. D. Smith, A. V. Turnbull and S. J. Y. Wang, J. Med. Chem., 2009, 52, 1558–1568 CrossRef CAS PubMed .
  121. H. Zhang, C. Shen, H.-R. Zhang, W.-X. Chen, Q.-Q. Luo and L. Ding, Mol. Diversity, 2021, 25, 1481–1495 CrossRef CAS PubMed .
  122. S. Cases, S. J. Smith, Y.-W. Zheng, H. M. Myers, S. R. Lear, E. Sande, S. Novak, C. Collins, C. B. Welchi, A. J. Lusisi, S. K. Erickson and R. V. Farese Jr, Proc. Natl. Acad. Sci. U. S. A., 1998, 95, 13018–13023 CrossRef CAS PubMed .
  123. J. Qi, W. Lang, E. Giardino, G. W. Caldwell, C. Smith, L. K. Minor, A. L. Darrow, G. Willemsens, K. DeWaepenaert, P. Roevens, J. T. M. Linders, Y. Liang and M. A. Connelly, J. Lipid Res., 2010, 51, 3559–3567 CrossRef CAS PubMed .
  124. J. H. Choi, J. N. Choi, S. Y. Lee, S. J. Lee, K. Kim and Y. K. Kim, Arch. Pharmacal Res., 2010, 33, 237–242 CrossRef CAS PubMed .
  125. M. Y. Chung, M.-C. Rho, J. S. Ko, S. Y. Ryu, K. H. Jeune, K. Kim, H. S. Lee and Y. K. Kim, Planta Med., 2004, 70, 258–260 CrossRef CAS PubMed .
  126. W. K. Oh, C.-H. Lee, J. H. Seo, M. Y. Chung, L. Cui, Z. T. Fomum, J. S. Kang and H. S. Lee, Arch. Pharmacal Res., 2009, 32, 43–47 CrossRef CAS PubMed .
  127. N. Tabata, M. Ito, H. Tomoda and S. Omura, Phytochemistry, 1997, 46, 683–687 CrossRef CAS PubMed .
  128. A. Casaschi, G. K. Maiyoh, B. K. Rubio, R. W. Li, K. Adeli and A. G. Theriault, J. Nutr., 2004, 134, 1340–1346 CrossRef CAS PubMed .
  129. H. Kondo, K. Hashizume, Y. Shibuya, T. Hase and T. Murase, Lipids, 2011, 46, 691–700 CrossRef CAS PubMed .
  130. J. S. Ko, M.-C. Rho, M. Y. Chung, H. Y. Song, J. S. Kang, K. Kim, H. S. Lee and Y. K. Kim, Planta Med., 2002, 68, 1131–1133 CrossRef CAS PubMed .
  131. J. S. Ko, S. Y. Ryu, Y. S. Kim, M. Y. Chung, J. S. Kang, M.-C. Rho, H. S. Lee and Y. K. Kim, Arch. Pharmacal Res., 2002, 25, 446–448 CrossRef CAS PubMed .
  132. N. T. Dat, X. F. Cai, M.-C. Rho, H. S. Lee, K. H. Bae and Y. H. Kim, Arch. Pharmacal Res., 2005, 28, 164–168 CrossRef CAS PubMed .
  133. J. Liu, X.-F. He, G.-H. Wang, E. F. Merino, S.-P. Yang, R.-X. Zhu, L.-S. Gan, H. Zhang, M. B. Cassera, H.-Y. Wang, D. G. I. Kingston and J.-M. Yue, J. Org. Chem., 2014, 79, 599–607 CrossRef CAS PubMed .
  134. L. Han, P. Lai and J.-R. Du, J. Evidence-Based Complementary Altern. Med., 2014, 823154 Search PubMed .
  135. M.-X. Xiu, Y.-M. Zhao, Y. Zhang, D.-X. Xiong, D. Wang, H.-S. Lee and L. Cui, Fitoterapia, 2021, 151, 104881 CrossRef CAS PubMed .
  136. B. W. Choi, H. S. Lee, K. B. Lee and B. H. Lee, Phytother. Res., 2011, 25, 1041–1045 CrossRef CAS PubMed .
  137. H. Tomoda, M. Ito, N. Tabata, R. Masuma, Y. Yamaguchi and S. Omura, J. Antibiot., 1995, 48, 937–941 CrossRef CAS PubMed .
  138. H. Tomoda, Y. Ohyama, T. Abe, N. Tabata, M. Namikoshi, Y. Yamaguchi, R. Masuma and S. Omura, J. Antibiot., 1999, 52, 689–694 CrossRef CAS PubMed .
  139. S. W. Lee, M.-C. Rho, J.-H. Choi, K. Kim, Y. S. Choi, H. S. Lee and Y. K. Kim, J. Microbiol. Biotechnol., 2008, 18, 1785–1788 CAS .
  140. Y.-M. Zhao, M.-X. Xiu, D. Wang, Y. Zhang, X.-Y. Zhang, X.-J. Shao, X.-H. Wang, H.-S. Lee and L. Cui, Phytochem. Lett., 2021, 44, 120–124 CrossRef CAS .
  141. A. Casaschi, B. K. Rubio, G. K. Maiyoh and A. G. Theriault, Atherosclerosis, 2004, 176, 247–253 CrossRef CAS PubMed .
  142. A. Casaschi, Q. Wang, K. Dang, A. Richards and A. Theriault, Lipids, 2002, 37, 647–652 CrossRef CAS PubMed .
  143. S. W. Lee, M.-C. Rho, H. R. Park, J.-H. Choi, J. Y. Kang, J. W. Lee, K. Kim, H. S. Lee and Y. K. Kim, J. Agric. Food Chem., 2006, 54, 9759–9763 CrossRef CAS PubMed .
  144. S. W. Lee, K. Kim, M.-C. Rho, M. Y. Chung, Y. H. Kim, S. Lee, H. S. Lee and Y. K. Kim, Planta Med., 2004, 70, 197–200 CrossRef CAS PubMed .
  145. N. Ugaki, D. Matsuda, H. Yamazaki, K. Nonaka, R. Masuma, S. Omura and H. Tomoda, J. Antibiot., 2012, 65, 15–19 CrossRef CAS PubMed .
  146. X. Wang, R. Zhao, W. Ji, J. Zhou, Q. Liu, L. Zhao, Z. Shen, S. Liu and B. Xu, ACS Med. Chem. Lett., 2022, 13, 118–127 CrossRef CAS PubMed .
  147. P. D. van Poelje, S. C. Potter and M. D. Erion, in Diabetes-Perspectives in Drug Therapy, ed. M. Schwanstecher, Springer-Verlag, Berlin, Heidelberg, 2011, chapter 12, pp. 279−302 Search PubMed .
  148. D. Sherpa, J. Chrustowicz, S. Qiao, C. R. Langlois, L. A. Hehl, K. V. Gottemukkala, F. M. Hansen, O. Karayel, S. von Gronau, J. R. Prabu, M. Mann, A. F. Alpi and B. A. Schulman, Mol. Cell, 2021, 81, 2445–2459 CrossRef CAS PubMed .
  149. R. Kaur, L. Dahiya and M. Kumar, Eur. J. Med. Chem., 2017, 141, 473–505 CrossRef CAS PubMed .
  150. J. Bie, S. Liu, Z. Li, Y. Mu, B. Xu and Z. Shen, Eur. J. Med. Chem., 2015, 90, 394–405 CrossRef CAS PubMed .
  151. G. Gomori, J. Biol. Chem., 1943, 148, 139–149 CrossRef CAS .
  152. S. Traniello, E. Melloni and S. Pontremoli, Arch. Biochem. Biophys., 1972, 149, 222–231 CrossRef CAS PubMed .
  153. F. Marcus, I. Edelstein and J. Rittenhouse, Biochem. Biophys. Res. Commun., 1984, 119, 1103–1108 CrossRef CAS PubMed .
  154. C. H. Fiske and J. Subbarow, J. Biol. Chem., 1925, 66, 375–400 CrossRef CAS .
  155. H. H. Tausky and E. A. Shorr, J. Biol. Chem., 1953, 202, 675–685 CrossRef .
  156. S. J. Pilkis, M. R. El-Maghrabi, J. Pilkis and T. Clau, J. Biol. Chem., 1981, 256, 3619–3622 CrossRef CAS PubMed .
  157. F. Mancini, J. Fiori, V. Cavrini and V. Andrisano, J. Sep. Sci., 2006, 29, 2395–2400 CrossRef CAS PubMed .
  158. M. R. El-Maghrabi, M. Gidh-Jain, L. A. R. Austin and S. J. Pilkis, J. Biol. Chem., 1993, 268, 9466–9472 CrossRef CAS PubMed .
  159. Y. Huang, B. Chi, Y. Xu, R. Song, L. Wei, L. Rao, L. Feng, Y. Ren and J. Wan, J. Mol. Graphics Modell., 2019, 86, 142–148 CrossRef CAS PubMed .
  160. Y. Huang, L. Wei, X. Han, H. Chen, Y. Ren, Y. Xu, R. Song, L. Rao, C. Su, C. Peng, L. Feng and J. Wan, Eur. J. Med. Chem., 2019, 184, 111749 CrossRef CAS PubMed .
  161. X. Han, Y. Huang, L. Wei, H. Chen, Y. Guo, Z. Tang, W. Hu, Q. Xia, Q. Wang, J. Yan and Y. Ren, Bioorg. Med. Chem., 2020, 28, 115624 CrossRef CAS PubMed .
  162. C. Proença, A. Oliveira, M. Freitas, D. Ribeiro, J. L. C. Sousa, M. J. Ramos, A. M. S. Silva, P. A. Fernandes and E. Fernandes, J. Nat. Prod., 2020, 83, 1541–1552 CrossRef PubMed .
  163. J. Revathy, S. Srinivasan, S. H. S. Abdullah and U. Muruganathan, Biomed. Pharmacother., 2018, 97, 98–106 CrossRef PubMed .
  164. L. Pari and S. Srinivasan, Biomed. Pharmacother., 2010, 64, 477–481 CrossRef CAS PubMed .
  165. Y. Zhang, X. Zhang, Z. Xiao, X. Zhang and H. Sun, Fitoterapia, 2022, 156, 105083 CrossRef CAS PubMed .
  166. R. Harini, M. Ezhumalai and K. V. Pugalendi, Eur. J. Pharmacol., 2012, 676, 89–94 CrossRef CAS PubMed .
  167. P. Vanithaa, C. Umaa, N. Suganyaa, E. Bhakkiyalakshmia, S. Suriyanarayananb, P. Gunasekaranc, S. Sivasubramanianc and K. M. Ramkumar, Environ. Toxicol. Pharmacol., 2014, 37, 326–335 CrossRef PubMed .
  168. P. Stanley Mainzen Prince and N. Kamalakkannan, J. Biochem. Mol. Toxicol., 2006, 20, 96–102 CrossRef CAS PubMed .
  169. Md. M. Alam, D. Meerza and I. Naseem, Life Sci., 2014, 109, 8–14 CrossRef CAS PubMed .
  170. S. Heng, K. M. Harris and E. R. Kantrowitz, Eur. J. Med. Chem., 2010, 45, 1478–1484 CrossRef CAS PubMed .
  171. S. Liu, M. J. Ammirati, X. Song, J. D. Knafels, J. Zhang, S. E. Greasley, J. A. Pfefferkorn and X. Qiu, J. Biol. Chem., 2012, 287, 13598–13610 CrossRef CAS PubMed .
  172. S. M. Sternisha and B. G. Miller, Arch. Biochem. Biophys., 2019, 663, 199–213 CrossRef CAS PubMed .
  173. K. Kamata, M. Mitsuya, T. Nishimura, J. I. Eiki and Y. Nagata, Structure, 2004, 12, 429–438 CrossRef CAS PubMed .
  174. E. Van Schaftingen, in Encyclopedia of Biological Chemistry: Third Edition, 2021, vol. 1, pp. 149–161 Search PubMed .
  175. J. Zhang, C. Li, T. Shi, K. Chen, X. Shen and H. Jiang, PLoS One, 2009, 4, e6304 CrossRef PubMed .
  176. S. Roy, M. V. Vega and N. J. Harmer, Catalysts, 2019, 9, 1–19 CrossRef CAS .
  177. G. Meisenberg and W. H. Simmons, Principles of Medical Biochemistry, 2017, pp. 392–411 Search PubMed .
  178. F. M. Ashcroft, M. Lloyd and E. A. Haythorne, Trends Endocrinol. Metab., 2023, 34, 119–130 CrossRef CAS PubMed .
  179. W. Li, X. Zhang, Y. Sun and Z. Liu, Pharmazie, 2020, 75, 230–235 CAS .
  180. K. S. Polonsky and C. F. Burant, Williams Textbook of Endocrinology, Elsevier, 2016, pp. 1385–1450 Search PubMed .
  181. T. J. Guzmán and C. M. Gurrola-Díaz, Arch. Physiol. Biochem., 2021, 127, 182–193 CrossRef PubMed .
  182. F. J. Bedoya, F. M. Matschinsky, T. Shimizu, J. J. O'Neil and M. C. Appel, J. Biol. Chem., 1986, 261, 10760–10764 CrossRef CAS PubMed .
  183. F. M. Matschinsky and H. Walk, Diabetes, 1990, 39, 647–652 CrossRef CAS PubMed .
  184. C. Hale, D. J. Lloyd, A. Pellacani and M. M. Véniant, Expert Opin. Ther. Targets, 2015, 19, 129–139 CrossRef CAS PubMed .
  185. A. Pautsch, N. Stadler, A. Löhle, W. Rist, A. Berg, L. Glocker, H. Nar, D. Reinert, M. Lenter, A. Heckel, G. Schnapp and S. G. Kauschke, Biochemistry, 2013, 52, 3523–3531 CrossRef CAS PubMed .
  186. F. M. Matschinsky and D. F. Wilson, Front. Physiol., 2019, 10, 148 CrossRef PubMed .
  187. R. Sarabu, S. J. Berthel, R. F. Kester and J. W. Tilley, Expert Opin. Ther. Pat., 2011, 21, 13–33 CrossRef CAS PubMed .
  188. K. S. Ashton, K. L. Andrews, M. C. Bryan, J. Chen, K. Chen, M. Chen, S. Chmait, M. Croghan, R. Cupples, C. Fotsch, J. Helmering, S. R. Jordan, R. J. M. Kurzeja, K. Michelsen, L. D. Pennington, S. F. Poon, G. Sivits, G. Van, S. L. Vonderfecht, R. C. Wahl, J. Zhang, D. J. Lloyd, C. Hale and D. J. S. Jean, J. Med. Chem., 2014, 57, 309–324 CrossRef CAS PubMed .
  189. F. J. Bedoya, R. Ramirez, E. Arilla and R. Goberna, Diabetes, 1984, 33, 858–863 CrossRef CAS PubMed .
  190. K. Chen, K. Michelsen, R. J. M. Kurzeja, J. Han, M. Vazir, D. J. St. Jean, C. Hale and R. C. Wahl, J. Biomol. Screening, 2014, 19, 1014–1023 CrossRef PubMed .
  191. V. V. Heredia, J. Thomson, D. Nettleton and S. Sun, Biochemistry, 2006, 45, 7553–7562 CrossRef CAS PubMed .
  192. M. Li, Y. Dang, Q. Li, W. Zhou, J. Zuo, Z. Yao, L. Zhang and G. Ji, Sci. Rep., 2019, 9, 1–12 CrossRef PubMed .
  193. Z. Song, H. Wang, L. Zhu, M. Han, Y. Gao, Y. Du and Y. Wen, Food Funct., 2015, 6, 461–469 RSC .
  194. A. Peter, N. Stefan, A. Cegan, M. Walenta, S. Wagner, A. Königsrainer, I. Königsrainer, F. Machicao, F. Schick, H. U. Häring and E. Schleicher, J. Clin. Endocrinol. Metab., 2011, 96, 1126–1130 CrossRef PubMed .
  195. C. Sanz, I. Roncero, P. Vázquez, M. A. Navas and E. Blázquez, J. Endocrinol., 2007, 193, 259–267 CAS .
  196. S. Sharma, K. Wadhwa, M. Choudhary and V. Budhwar, Nat. Prod. Res., 2022, 36, 2962–2976 CrossRef CAS PubMed .
  197. K. O. Faloye, B. D. Bekono, E. G. Fakola, M. D. Ayoola, O. I. Bello, O. G. Olajubutu, O. D. Owoseeni, S. Mahmud, M. Alqarni, A. A. Al Awadh, M. M. Alshahrani and A. J. Obaidullah, Molecules, 2021, 26, 7211 CrossRef CAS PubMed .
  198. S. Chandrasekaran, N. Ramajayam and P. Pachaiappan, Biomed. Pharmacother., 2018, 103, 539–545 CrossRef CAS PubMed .
  199. I. Dhananjayan, S. Kathiroli, S. Subramani and V. Veerasamy, Biomed. Pharmacother., 2017, 88, 1069–1079 CrossRef CAS PubMed .
  200. Z. Y. Hao, Y. G. Cao, Y. Wang, C. L. Zhang, H. Luo, D. Liang, Y. F. Liu, R. Y. Chen and D. Q. Yu, Phytochem. Lett., 2021, 45, 37–43 CrossRef CAS .
  201. R. Harini and K. V. Pugalendi, J. Basic Clin. Physiol. Pharmacol., 2010, 21, 79–92 Search PubMed .
  202. C. Su, C. Yang, M. Gong, Y. Ke, P. Yuan, X. Wang, M. Li, X. Zheng and W. Feng, Molecules, 2019, 24, 1–14 Search PubMed .
  203. R. Harini, M. Ezhumalai and K. V. Pugalendi, Eur. J. Pharmacol., 2012, 676, 89–94 CrossRef CAS PubMed .
  204. P. Daisy, K. Balasubramanian, M. Rajalakshmi, J. Eliza and J. Selvaraj, Phytomedicine, 2010, 17, 28–36 CrossRef CAS PubMed .
  205. D. Prabakaran and N. Ashokkumar, J. Funct. Foods, 2012, 4, 776–783 CrossRef CAS .
  206. R. Murali, S. Srinivasan and N. Ashokkumar, Biochimie, 2013, 95, 1848–1854 CrossRef CAS PubMed .
  207. R. Jayaraman, S. Subramani, S. H. Sheik Abdullah and M. Udaiyar, Biomed. Pharmacother., 2018, 97, 98–106 CrossRef CAS PubMed .
  208. R. Sundaram, E. Nandhakumar and H. Haseena Banu, Toxicol. Mech. Methods, 2019, 29, 644–653 CrossRef CAS PubMed .
  209. H. Alkhalidy, W. Moore, Y. Wang, J. Luo, R. P. McMillan, W. Zhen, K. Zhou and D. Liu, Molecules, 2018, 23, 2338 CrossRef PubMed .
  210. R. Sundaram, P. Shanthi and P. Sachdanandam, Phytomedicine, 2014, 21, 793–799 CrossRef CAS PubMed .
  211. R. Chandramohan, L. Pari, A. Rathinam and B. A. Sheikh, Chem.-Biol. Interact., 2015, 229, 44–54 CrossRef CAS PubMed .
  212. R. Murali and R. Saravanan, Biomed. Prev. Nutr., 2012, 2, 269–275 CrossRef .
  213. S. Babukumar, V. Vinothkumar, C. Sankaranarayanan and S. Srinivasan, Pharm. Biol., 2017, 55, 1442–1449 CrossRef CAS PubMed .
  214. B. A. Sheikh, L. Pari, A. Rathinam and R. Chandramohan, Biochimie, 2015, 112, 57–65 CrossRef CAS PubMed .
  215. R. Subramanian, M. Z. Asmawi and A. Sadikun, Pharm. Biol., 2008, 46, 772–780 CrossRef CAS .
  216. R. Saravanan, K. V. Babu and V. Ramachandran, J. Physiol. Biochem., 2012, 68, 421–431 CrossRef CAS PubMed .
  217. G. Ni, Z. F. Shen, Y. Lu, Y. H. Wang, Y. B. Tang, R. Y. Chen, Z. Y. Hao and D. Q. Yu, J. Org. Chem., 2011, 76, 2056–2061 CrossRef CAS PubMed .
  218. P. Kalaiarasi and K. V. Pugalendi, Eur. J. Pharmacol., 2009, 606, 269–273 CrossRef CAS PubMed .
  219. V. Ramachandran and R. Saravanan, Phytomedicine, 2013, 20, 230–236 CrossRef CAS PubMed .
  220. S. Vikhe, R. Kunkulol and D. Raut, J. Ayurveda Integr. Med., 2022, 13, 100618 CrossRef CAS PubMed .
  221. P. S. Ngubane, B. Masola and C. T. Musabayane, Renal Failure, 2011, 33, 434–439 CrossRef CAS PubMed .
  222. R. M. Perez-Gutierrez and M. Damian-Guzman, Biol. Pharm. Bull., 2012, 35, 1516–1524 CrossRef CAS PubMed .
  223. G. Chandramohan, S. Ignacimuthu and K. V. Pugalendi, Eur. J. Pharmacol., 2008, 590, 437–443 CrossRef CAS PubMed .
  224. L. Y. Tian, X. Bai, X. H. Chen, J. B. Fang, S. H. Liu and J. C. Chen, Phytomedicine, 2010, 17, 533–539 CrossRef CAS PubMed .
  225. Q. Min, X. Cai, W. Sun, F. Gao, Z. Li, Q. Zhang, L. S. Wan, H. Li and J. Chen, Sci. Rep., 2017, 7, 1–9 CrossRef PubMed .
  226. V. Kumar, P. C. Bhatt, G. Kaithwas, M. Rashid, F. A. Al-abbasi, J. A. J. Khan, F. Anwar and A. Verma, Beni-Suef Univ. J. Basic Appl. Sci., 2016, 5, 255–276 Search PubMed .
  227. H. He, W. G. Yu, J. P. Yang, S. Ge and Y. H. Lu, J. Agric. Food Chem., 2016, 64, 2475–2484 CrossRef CAS PubMed .
  228. S. Bhattacharya, N. Oksbjerg, J. F. Young and P. B. Jeppesen, Diabetes, Obes. Metab., 2014, 16, 602–612 CrossRef CAS PubMed .
  229. Y. Tang and A. Chen, Br. J. Pharmacol., 2010, 161, 1137–1149 CrossRef CAS PubMed .
  230. Z. Yang, L. Zhang, J. Liu, F. Lu, L. Wang, Y. Chen and D. Li, BMC Complementary Altern. Med., 2019, 19, 1–9 CrossRef PubMed .
  231. J. Wang, Y. Huang, K. Li, Y. Chen, D. Vanegas, E. S. McLamore and Y. Shen, PLoS One, 2016, 11, 1–24 Search PubMed .
  232. Y. Ou, Z. Ren, J. Wang and X. Yang, Chem.-Biol. Interact., 2016, 247, 49–54 CrossRef CAS PubMed .
  233. J. Hao, C. Chen, K. Huang, J. Huang, J. Li, P. Liu and H. Huang, Eur. J. Pharmacol., 2014, 745, 152–165 CrossRef CAS PubMed .
  234. J. Lee, H. I. Lee, K. Il Seo, H. W. Cho, M. J. Kim, E. M. Park and M. K. Lee, Indian J. Exp. Biol., 2014, 52, 683–691 Search PubMed .
  235. A. S. Grewal, N. Sharma and S. Singh, Int. J. Pharm. Qual., 2019, 10, 571–577 Search PubMed .
  236. A. S. Grewal, S. Arora, N. Sharma and S. Singh, Plant Arch., 2020, 20, 3768–3771 Search PubMed .
  237. Q. Xiao, J. J. Jackson, A. Basak, J. M. Bowler, B. G. Miller and A. Zakarian, Nat. Chem., 2013, 5, 410–416 CrossRef CAS PubMed .
  238. M. K. Hasan, I. Ara, M. S. A. Mondal and Y. Kabir, Heliyon, 2021, 7, e07240 CrossRef CAS PubMed .
  239. R. Sundaram, P. Shanthi and P. Sachdanandam, Phytomedicine, 2014, 21, 793–799 CrossRef CAS PubMed .
  240. N. Schormann, K. L. Hayden, P. Lee, S. Banerjee and D. Chattopadhyay, Protein Sci., 2019, 28, 1771–1784 CrossRef CAS PubMed .
  241. M. Khalid, J. Alkaabi, M. A. B. Khan and A. Adem, Int. J. Mol. Sci., 2021, 22, 8590 CrossRef CAS PubMed .
  242. C. H. Trinh, A. Asipu, D. T. Bonthron and S. E. V. Phillips, Acta Crystallogr., Sect. D: Biol. Crystallogr., 2009, 65, 201–211 CrossRef CAS PubMed .
  243. D. T. Bonthron, N. Brady, L. A. Donaldson and B. Steinmann, Hum. Mol. Genet., 1994, 3, 1627–1631 CrossRef CAS PubMed .
  244. T. Ishimoto, M. A. Lanaspa, M. P. T. Le, G. E. Garcia, C. P. Diggle, P. S. MacLean, M. R. Jackman, A. Asipu, C. A. Roncal-Jimenez, T. Kosugia, C. J. Rivarda, S. Maruyamad, B. Rodriguez-Iturbe, L. G. Sánchez-Lozada, D. T. Bonthron, Y. Y. Sauting and R. J. Johnson, Proc. Natl. Acad. Sci. U. S. A., 2012, 109, 4320–4325 CrossRef CAS PubMed .
  245. X. Zhang, F. Song, G. H. Kuo, A. Xiang, A. C. Gibbs, M. C. Abad, W. Sun, L. C. Kuo and Z. Sui, Bioorg. Med. Chem. Lett., 2011, 21, 4762–4767 CrossRef CAS PubMed .
  246. K. Huard, K. Ahn, P. Amor, D. A. Beebe, K. A. Borzilleri, B. A. Chrunyk, S. B. Coffey, Y. Cong, E. L. Conn, J. S. Culp, M. S. Dowling, M. F. Gorgoglione, J. A. Gutierrez, J. D. Knafels, E. A. Lachapelle, J. Pandit, K. D. Parris, S. Perez, J. A. Pfefferkorn, D. A. Price, B. Raymer, T. T. Ross, A. Shavnya, A. C. Smith, T. A. Subashi, G. J. Tesz, B. A. Thuma, M. Tu, J. D. Weaver, Y. Weng, J. M. Withka, G. Xing and T. V. Magee, J. Med. Chem., 2017, 60, 7835–7849 CrossRef CAS PubMed .
  247. P. Jegatheesan and J. P. De Bandt, Nutrients, 2017, 9, 1–13 CrossRef PubMed .
  248. C. Zhang, L. Li, Y. Zhang and C. Zeng, Biomed. Pharmacother., 2020, 131, 110795 CrossRef CAS PubMed .
  249. M. P. T. Le, M. A. Lanasp|a, C. M. Cicerchi, J. Rana, J. D. Scholten, B. L. Hunter, C. J. Rivard, R. K. Randolph and R. J. Johnson, PLoS One, 2016, 11, 1–17 Search PubMed .
  250. B. E. Maryanoff, J. C. O'Neill, D. F. McComsey, S. C. Yabut, D. K. Luci, A. D. Jordan, J. A. Masucci, W. J. Jones, M. C. Abad, A. C. Gibbs and I. Petrounia, ACS Med. Chem. Lett., 2011, 2, 538–543 CrossRef CAS PubMed .
  251. R. C. Adelman, F. J. Ballard and S. Weinhouse, J. Biol. Chem., 1967, 242, 3360–3365 CrossRef CAS PubMed .
  252. M. Wang, L. Lin, J. J. Lu and X. Chen, Pharmacol. Res., 2021, 165, 105483 CrossRef CAS PubMed .
  253. H. J. Liang, F. M. Suk, C. K. Wang, L. F. Hung, D. Z. Liu, N. Q. Chen, Y. C. Chen, C. C. Chang and Y. C. Liang, Chem.-Biol. Interact., 2009, 181, 309–315 CrossRef CAS PubMed .
  254. O. D. Alabi, S. M. Gunnink, B. D. Kuiper, S. A. Kerk, E. Braun and L. L. Louters, Life Sci., 2014, 102, 105–110 CrossRef CAS PubMed .
  255. V. Kumar, P. C. Bhatt, G. Kaithwas, M. Rashid, F. A. Al-abbasi, J. A. J. Khan, F. Anwar and A. Verma, Beni-Suef Univ. J. Basic Appl. Sci., 2016, 5, 255–276 Search PubMed .
  256. S. P. Chen, S. R. Lin, T. H. Chen, H. S. Ng, H. S. Yim, M. K. Leong and C. F. Weng, Biomed. Pharmacother., 2021, 144, 112333 CrossRef CAS PubMed .
  257. K. Huard, K. Ahn, P. Amor, D. A. Beebe, K. A. Borzilleri, B. A. Chrunyk, S. B. Coffey, Y. Cong, E. L. Conn, J. S. Culp, M. S. Dowling, M. F. Gorgoglione, J. A. Gutierrez, J. D. Knafels, E. A. Lachapelle, J. Pandit, K. D. Parris, S. Perez, J. A. Pfefferkorn, D. A. Price, B. Raymer, T. T. Ross, A. Shavnya, A. C. Smith, T. A. Subashi, G. J. Tesz, B. A. Thuma, M. Tu, J. D. Weaver, Y. Weng, J. M. Withka, G. Xing and T. V. Magee, J. Med. Chem., 2017, 60, 7835–7849 CrossRef CAS PubMed .
  258. A. Anighoro, J. Bajorath and G. Rastelli, J. Med. Chem., 2014, 57, 7874–7887 CrossRef CAS PubMed .
  259. Y. Hu and J. Bajorath, Drug Discovery Today, 2013, 18, 644–650 CrossRef CAS PubMed .
  260. C. L. Chai and P. Mátyus, Future Med. Chem., 2016, 8, 29–38 CrossRef CAS PubMed .
  261. J. B. Baell and J. W. M. Nissink, ACS Chem. Biol., 2018, 13, 36–44 CrossRef CAS PubMed .
  262. A. Moya-García, T. Adeyelu, F. A. Kruger, N. L. Dawson, J. G. Lees, J. P. Overington, C. Orengo and J. A. Raena, Sci. Rep., 2017, 7, 10102 CrossRef PubMed .
  263. R. Jansson-Löfmark, S. Hjorth and J. Gabrielsson, Clin. Pharmacol. Ther., 2020, 108, 298–305 CrossRef PubMed .

This journal is © The Royal Society of Chemistry 2023