Introduction
Hyperlipidemia is a pathological dysfunction of lipid metabolism that has varied causes. The scientific manifestations of hyperlipidemia embody elevated serum ldl cholesterol, triglycerides, and low-density lipoprotein ldl cholesterol (LDL-C), and decreased serum high-density lipoprotein ldl cholesterol (HDL-C). Imbalance of LDL-C and HDL-C can enhance the chance of cardiovascular (CV) occasions, together with myocardial infarction and stroke.1 Information launched by the American Coronary heart Affiliation in 2018 confirmed that CV illness (CVD) is probably the most deadly illness worldwide.2 Based on the 2010 international burden of illness research, 15.6 million folks died of CVD, accounting for 29.6% of all deaths.3 Hyperlipidemia has a protracted illness course, and plenty of underlying causes, reflecting its complicated etiology.4 Dyslipidemia is a major danger issue for coronary artery illness and stroke; therefore, prevention and applicable administration of dyslipidemia can markedly decrease the associated morbidity and mortality.5 Holven et al described the significance of early identification and remedy of sufferers with familial hypercholesterolemia for lowering ldl cholesterol burden and danger of CHD.6 Lipid-lowering remedy is a cornerstone of CV danger modification methods, which may scale back LDL-C by 30–50% and proportionally scale back CV occasions.7 Though statins are typically effectively tolerated, they don’t seem to be all the time ample to realize LDL-C objectives for a lot of sufferers, and trigger quite a few particular destructive results on muscle, liver, and kidney, in addition to rising the chance of new-onset diabetes mellitus and hemorrhagic stroke.8,9 Subsequently, there’s an pressing have to develop new highly effective medication to deal with hyperlipidemia and scale back the harms to human well being and longevity worldwide related to CV occasions.
Pure merchandise can facilitate a multi-component, multi-target, multi-channel, and multi-dimensional total remedy networks.10 Thus, pure merchandise symbolize various sources for creating new medication, with greater effectivity, higher security, and fewer unwanted effects.11 Furthermore, pure merchandise can play important roles in stopping and treating many human illnesses, together with CVDs, metabolic syndromes, most cancers, diabetes, weight problems, and neurological issues.12,13
Diosgenin, a well known steroid sapogenin, happens broadly in varied medical crops,14 and is especially remoted from Dioscoreaceae, Agavaceae, Amaryllidaceae, Liliaceae, Solanaceae, Scrophulariaceae, Amaryllidaceae, Leguminosae, and Rhamnaceae.15 The perfect-known supply of varied steroidal saponins, together with spirostane and furostane sorts, is fenugreek (Trigonella foenum graecum) seeds.16 Lately, diosgenin has attracted rising consideration attributable to its efficacy in treating varied metabolic illnesses, together with diabetes,17 CVDs,18 neurological illness,19 osteoporosis,20 and hyperlipidemia,21 along with anti-cancer results, that are mediated by way of a number of targets and regulate varied indicators.22,23 Quite a few animal experiments and scientific trials have proven that diosgenin can scale back blood lipids by reducing plasma low-density lipoprotein (LDL) and rising high-density lipoprotein (HDL). Ldl cholesterol reducing drug cholestyramine can mix with bile acid (BA) to type insoluble complicated and stop its reabsorption. In contrast with the affect of cholestyramine, diosgenin can intrude with the absorption of exogenous and endogenous ldl cholesterol, promote the secretion of ldl cholesterol into bile, and enhance the excretion of impartial sterols with out affecting the bile and fecal excretion of bile acid.24 The impact of diosgenin mixed with clofibrate in lowering plasma LDL is extra sturdy than that of a single drug; therefore, diosgenin will be mixed with different lipid-lowering medicines to boost their results.25 Additional, diosgenin has demonstrated excellent potential for reducing lipids in scientific trials and animal experiments and represents a brand new avenue of exploration for attaining lipid discount.
On this paper, we elaborate the detailed mechanism underlying lipid metabolism regulation by diosgenin. As proven in Figure 1, this consists of the results of diosgenin in inhibiting intestinal lipid absorption, regulating ldl cholesterol transport, selling ldl cholesterol conversion into bile acid and its excretion, inhibiting endogenous lipid biosynthesis, antioxidation results, regulating lipoprotein lipase exercise, and regulating transcription components associated to lipid metabolism.
Physicochemical Properties of Diosgenin
Diosgenin (3β-Hydroxy-5-spirostene) (Figure 2) is a C27 spironosteroidal saponin of the spirosterol steroid household that participates in varied physiological and biochemical actions. Diosgenin is structurally just like ldl cholesterol and different steroids, with molecular components C27H42O3, density 1.1 ± 0.1g/cm3, and relative molecular mass 414.63. Diosgenin is the first precursor of varied pharmacologically lively steroids, together with corticosteroids and oral contraceptives and takes the type of a white acicular crystal or gentle amorphous powder, which is thermally and chemically secure underneath varied bodily situations. The melting level of diosgenin is above 200°C and the compound is comparatively secure underneath gentle, however unstable underneath hydrochloric acid, which causes its speedy decomposition.26 Diosgenin has low solubility, attributable to its robust hydrophobicity; its solubility in water is roughly 0.7 ng/mL.27 However, it’s extremely soluble in most nonpolar natural solvents (akin to chloroform, dichloroethane, propanol, ethyl acetate, and propyl acetate) and a few polar solvents (together with acetone, methanol, and anhydrous ethanol).28
Determine 2 Chemical construction of diosgenin. Its molecular components is C27H42O3, density is 1.1±0.1g/cm3, and relative molecular mass is 414.63. |
Sources of Diosgenin
Diosgenin is a pure steroidal saponin, which is derived from the hydrolysis of dioscin. Diosgenin will be commercially extracted from a number of crops, together with Trigonella,29 Costus,30 Aletris, Smilax,31 and plenty of species of Dioscorea.32 The first supply of diosgenin is fenugreek seeds.29 The content material of saponins in fenugreek seeds is 169 g/kg. After hydrolysis for 1 h, the saponins are fully eliminated and sapogenin launch is maximal. Extracts of hydrolyzed fenugreek and quinoa comprise the very best fraction of sapogenin and minor fractions of phytosterol and tocopherol.33 Diosgenin is a vital materials for the synthesis of steroid hormone medicines. For instance, dehydroepiandrosterone will be produced semi-synthetically from pure precursors, primarily diosgenin.34 Historically, diosgenin has been extracted from the rhizome of Dioscorea zingiberensis C. H. Wright by acid hydrolysis. Extra lately, a brand new technique of direct penicillin biotransformation, that’s environmental-friendly, easy, and energy-saving, has been developed and is taken into account a possible substitute for acid hydrolysis within the diosgenin extraction trade.35
The Pharmacological Mechanism Underlying the Use of Diosgenin for Remedy of Hyperlipidemia
Diosgenin has quite a few pharmacological results, together with anti-tumor exercise, bettering CV operate, regulating immunity, inflicting anti-platelet aggregation, and reducing blood lipids.22 Lately, the lipid-regulating operate of diosgenin has attracted rising consideration amongst researchers. In people, lipid homeostasis is regulated by homeostatic intestinal absorption mechanisms, endogenous lipid biosynthesis and metabolism, ldl cholesterol transport, ldl cholesterol conversion to bile acid, and biliary excretion. The metabolic enzymes and receptors concerned in lipid metabolism are regulated by varied transcription components. The operate of diosgenin in treating hypolipidemia via regulating the dynamics of various elements of the metabolic cycle is illustrated in Figure 3. The experimental design, pharmacological proof, and potential mechanism of diosgenin towards hyperlipidemia are summarized in Table 1.
Desk 1 The Impact of Diosgenin on Markers Related to Lipid Metabolism |
Inhibition of Intestinal Lipid Absorption
Intestinal absorption from the food plan is a vital supply of serum lipids. Ldl cholesterol absorption is a key regulation goal of lipid metabolism in people, as a result of it determines the quantity of endogenous bile and ldl cholesterol retained within the food plan, thus affecting ldl cholesterol homeostasis. Fenugreek seeds containing diosgenin induced a major discount of plasma ldl cholesterol in diabetic canine, related to interference in ldl cholesterol absorption.36 Additional, an in vivo experiment confirmed that intraperitoneal injection of 0.5 mg/kg protodioscin, a steroidal saponin just like diosgenin from Dioscorea nipponica rhizomes, may considerably scale back blood ldl cholesterol by 40% in male Sprague–Dawley (SD) rats fed a high-fat food plan (HFD).37 Researchers additionally investigated the inhibitory impact of diosgenin on lipid absorption utilizing experiments on cynomolgus macaques. The macaques have been fed a semi-purified food plan containing 0.1% ldl cholesterol or an identical food plan containing 1% diosgenin throughout two 3-week intervals, and acquired a typical chow food plan for five weeks between the 2 experimental intervals. The outcomes confirmed that diosgenin lowered ldl cholesterol from 292 to 172mg/dl and intestinal absorption of exogenous ldl cholesterol from 62.4% to 26.0%, in addition to rising internet endogenous ldl cholesterol secretion from –0.8 to 93.5 mg/day.38 Niemann-Choose C1-Like 1 (NPC1L1) is a polytopic transmembrane protein with a crucial function in ldl cholesterol absorption. Blocking NPC1L1 endocytosis considerably reduces ldl cholesterol endocytosis39 and hepatic NPC1L1 has a direct function in regulating bile ldl cholesterol excretion and hepatic/blood levels of cholesterol.40 Expression of NPC1L1 within the 0.15 or 0.3 g/kg diosgenin group confirmed a dose-dependent lower in contrast with the HFD group, demonstrating that diosgenin may scale back intestinal absorption by inhibiting NPC1L1.21 Each diosgenin and complete saponins of yellow yam can considerably scale back blood ldl cholesterol content material, the place absolutely the dose of diosgenin contains solely half of the entire saponins in yellow yam. The anti-hypercholesterolemia impact of diosgenin was superior to that of complete saponins from yellow yam, doubtless as a result of the polarity and spatial construction of diosgenin are similar to these of ldl cholesterol, suggesting that diosgenin can doubtless be dispersed into metacolloidals by way of bile acid exercise, after which absorbed instantly by the gut. When diosgenin and ldl cholesterol are each current, diosgenin competes for bile acid binding to inhibit ldl cholesterol absorption.41 Total, these outcomes point out that diosgenin can inhibit lipid absorption by downregulating NPC1L1 expression and competing with ldl cholesterol for bile acids within the intestinal tract.
Regulation of Ldl cholesterol Transport
Along with blood ldl cholesterol and triglycerides, lipoproteins, akin to HDL and LDL, even have essential roles in lipid metabolism, as they operate to move lipids and ldl cholesterol which can be typically insoluble in blood. Kuang et al42 utilized diosgenin to the lipogenic regular human liver (L02) cell mannequin, and the outcomes confirmed that diosgenin may up-regulate expression of Caveolin-1 protein, which is carefully associated to ldl cholesterol transport, and scale back intracellular levels of cholesterol. Protodioscin has potent HDL and LDL (particularly LDL) reducing results.37 Quite a few research have proven that HDL-C focus is inversely correlated with CVD danger; therefore, elevating plasma HDL-C ranges could defend towards CVD.43 In rats fed a high-cholesterol food plan (HCD), supplementation with 0.5/100 g diosgenin for 4 weeks led to a slight enhance in serum HDL-C and fecal bile acid ranges, and a lower in lipid droplet measurement.44 Sterol regulatory element-binding proteins (SREBPs) regulate the steadiness of LDL, HDL, and triglyceride ranges in vivo. In human THP-1 macrophages, methyl protodioscin (MPD) inhibits SREBP1c and SREBP2 transcription and reduces ranges of microRNA 33a/b, which is encoded in SREBP gene introns, resulting in a corresponding enhance in ATP binding cassette A1 (ABCA1) ranges.45 Since ABCA1 is a key protein in HDL biogenesis, low miR-33a/b ranges end in excessive HDL-C. Additional, MPD additionally decreases expression of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGCR), fatty acid synthase (FAS), and acyl-CoA carboxylase (ACC) genes, that are concerned in ldl cholesterol and fatty acid synthesis. Subsequently, MPD could enhance HDL-C ranges, and reduce LDL-C and triglyceride ranges.
Scavenger receptor class B sort I (SRB1) is a receptor physiologically associated to HDL, which participates in reverse transport of ldl cholesterol (RCT) and helps to move extra ldl cholesterol to the liver within the type of ldl cholesterol ester (CE), after which excrete it into bile and feces.46 Carboxylesterase 1 (CES-1) reveals cholesteryl ester hydrolase exercise, and contributes to hydrolyzation of HDL-CE to free ldl cholesterol (FC), which is accessible for bile acid synthesis.47 Ldl cholesterol 7α-hydroxylase (CYP7A1) is the preliminary and rate-limiting enzyme within the classical BA artificial pathway, which regulates the entire charge of BA era. Farnesol X receptor (FXR) is a nuclear receptor with elementary roles in sustaining BA homeostasis and in sustaining the steadiness between ldl cholesterol and BA. BAs, particularly hydrophobic BAs, act as ligands for FXR and put it to use of their destructive suggestions loop.48 In an in vivo experiment, Yu et al49 demonstrated that diosgenin (150 or 300 mg/kg/d) can scale back physique weight and blood lipid ranges of rats fed a HFD, and considerably enhance liver steatosis and intestinal construction. Additional, diosgenin can enhance the expression of SRB1, CES-1, and CYP7A1 in rat liver and inhibit FXR-mediated sign transduction, in addition to rising SRB1 and CES-1 expression in rat gut, inhibiting ldl cholesterol absorption, and selling RCT. Therefore, diosgenin promotes RCT and ldl cholesterol clearance via the SRB1/CES-1/CYP7A1/FXR pathway, which suggests potential new approaches to alleviate hypercholesterolemia. Collectively, the outcomes above point out that diosgenin can enhance HDL-C and reduce LDL-C and really low-density lipoprotein (VLDL) ldl cholesterol (VLDL-C) ranges by regulating ldl cholesterol transport, and thus take part in regulating blood lipid ranges.
Inhibiting Endogenous Lipid Biosynthesis
HMGCR is an endoplasmic reticulum sure peroxisome enzyme that catalyzes the discount of HMG-CoA to COA and mevalonate, which is a rate-limiting response within the de novo ldl cholesterol biosynthesis pathway; it’s extremely expressed within the liver and performs a central function within the regulation of ldl cholesterol metabolism.50 It’s established that HMGCR inhibitors can successfully scale back plasma levels of cholesterol in most animals, together with people and these inhibitors are broadly used as statins to cut back levels of cholesterol by inhibiting ldl cholesterol synthesis.51 Hao et al confirmed that oral administration of diosgenin (40 mg/kg orally for 45 days) may considerably scale back mRNA ranges encoding HMGCR within the liver of diabetic rats, suggesting that diosgenin can considerably scale back ldl cholesterol synthesis.52 Diosgenin could inhibit ldl cholesterol biosynthesis by blocking the substrate from getting into the lively website of the HMGCR enzyme. Uemura et al reported that diosgenin (5 and 10 mmol/l) decreased triglyceride content material and mRNA expression ranges of genes concerned in lipid synthesis (FAS, stearoyl-CoA desaturase 1 (SCD-1), and ACC) by inhibiting SREBP-1c mRNA expression, leading to inhibited lipid accumulation in hepatocellular carcinoma (HepG2) cells.53 A research to research the lipid-lowering results of various granule therapies (50 g/kg 50 mesh-size flour or 50 g/kg nanoscale flour) confirmed that each Dioscorea pseudojaponica particle sorts may activate adenosine 5ʹ-monophosphate-activated protein kinase (AMPK) and inactivate ACC, as demonstrated by elevated ranges of phosphorylated enzyme.54 AMPK activation will increase the speed of the catabolic (ATP manufacturing) pathway and reduces the speed of the anabolic (ATP utilization) pathway.55 Cheng hypothesized that diosgenin can stimulate AMPK activation, promote free fatty acid (FFA) decomposition and, underneath the synergistic impact of liver X receptor alpha (LXRα), inhibit SREBPIC and scale back triglyceride synthesis.56 Poudel et al demonstrated that diosgenin targets the AMPK/MAPK pathway, suppresses mitotic clonal growth through the early section of adipogenesis, and reduces the expression of adipogenic transcription components in 3T3-L1 cells throughout adipogenesis. At a dose of 4 µM, diosgenin inhibited lipid accumulation in 3T3-L1 cells, however didn’t have an effect on cell viability. Additional, diosgenin may regulate weight and fats accumulation induced by HFD in overweight mice.57 In abstract, diosgenin inhibits one or a number of hyperlinks within the ldl cholesterol and triglyceride biosynthetic pathways, thereby lowering ldl cholesterol and triglyceride ranges.
Selling Conversion of Ldl cholesterol into Bile Acid and Excretion
The classical route for the human physique to take away ldl cholesterol lipid is by way of promotion of ldl cholesterol excretion into biliary ldl cholesterol, the conversion of ldl cholesterol into bile acid, and its subsequent fecal excretion.58,59 The ATP-binding cassette (ABC) transporters, G5 (ABCG5) and G8 (ABCG8), are hemi-transporters, which play an essential function in stopping the buildup of dietary sterols (together with ldl cholesterol and phytosterols) within the physique.60 Li et al reported that, after remedy with 0.15 or 0.3 g/kg diosgenin, ABCG5/G8 expression was elevated within the liver and gut of HFD rats, suggesting that it will probably promote bile ldl cholesterol secretion.21 Temel et al61 confirmed that fecal excretion of impartial sterols (ldl cholesterol and its bacterial metabolites) in wild-type mice fed with 0.1% diosgenin elevated from 4.2 to 52 µmol/day/100 g physique weight. In contrast with wild-type mice receiving a management food plan, the fecal impartial sterol excretion of NPC1L1 knockout mice was additionally considerably elevated, from 63 to 140 µmol/day/100 g physique weight. This research demonstrated that diosgenin promotes fecal ldl cholesterol excretion independently of NPC1L1-mediated ldl cholesterol absorption, and that diosgenin could facilitate fecal ldl cholesterol loss to a better extent in NPC1L1 knockout mice than in wild-type mice.
ABCA1 mediates the switch of mobile phospholipids (PL) and FC to apolipoprotein A-I (ApoA-I) and associated proteins in extracellular medium, performing as a lipid transporter.62 Diosgenin can promote ABCA1 expression in macrophages by inhibiting miR-19b ranges, and speed up macrophage ldl cholesterol efflux via the ABCA1 pathway and RCT course of.63 In HepG2 cells, 25 µm pseudoprotodioscin (PPD), a steroidal saponin just like diosgenin, considerably elevated ranges of ABCA1 protein and mRNA, and promoted ApoA-1 mediated ldl cholesterol excretion. The underlying mechanism entails PPD inhibition of SREBP1c and SREBP2 transcription by lowering microRNA33a/b ranges, resulting in elevated ABCA1.64 In abstract, diosgenin can promote the conversion of ldl cholesterol to bile acid and enhance its excretion in varied methods; nevertheless, particulars of the complicated molecular mechanisms concerned stay to be clarified.
Oxidative Stress and Anti-Oxidant Properties
Oxidation-reduction equilibrium is crucial to keep up the conventional operation of significant cell capabilities. Oxidative stress is frequent in some illnesses, together with most cancers, CVD, atherosclerosis, and diabetes.65 Endogenous markers of oxidative stress embody superoxide dismutase (SOD), glutathione peroxidase (GPx), and lowered glutathione (GSH). Throughout oxidative stress, phospholipids in cell membranes are oxidized by thiobarbituric acid reactive substances (TBARS), which hinder their organic capabilities. Diosgenin has potent in vitro anti-oxidative and in vivo anti-inflammatory actions66 and participates in glucose metabolism, reduces oxidative stress, will increase cell viability, and reduces reactive oxygen species ranges.67 Sangeetha et al68 assessed the antioxidant standing of experimental rat liver and coronary heart tissue samples handled with and with out diosgenin. The rats have been fed with an HFD for 8 weeks after which acquired streptozotocin (STZ) injection to induce a kind 2 diabetes mannequin. GSH ranges have been greater in animals handled with diosgenin (40 mg/kg, 80 mg/kg), though they didn’t attain the vary of these in regular rats. Diosgenin may preserve SOD and GPX actions and management TBARS manufacturing. Rats with power renal failure (CRF) have been administered diosgenin orally (40 mg/kg/day), and subsequent outcomes demonstrated that diosgenin elevated GSH ranges and restored endothelial nitric oxide synthase (eNOS) mRNA expression. Additional, diosgenin inhibits dyslipidemia and angiotensin changing enzyme (ACE) exercise attributable to CRF. Total, information revealed so far present that diosgenin has potential to guard blood vessels from oxidative stress and dyslipidemia.69 Xu et al70 discovered that dioscin considerably attenuated the rise in malondialdehyde ranges attributable to ethanol and considerably elevated SOD, GSH, GSH-Px, and GSR ranges. These information point out that dioscin has a superb protecting impact towards ethanol-induced liver harm by lowering oxidative stress, inflammatory cytokine manufacturing, apoptosis, and liver steatosis. Additional, diosgenin acts as an antioxidant in cell membranes, which can present safety towards oxidative injury of polyunsaturated fatty acids.71 Fenugreek, which incorporates diosgenin, can enhance glucose metabolism dysfunction attributable to HFD by selling adipocyte differentiation, inhibiting irritation of white adipose tissue, and miniaturizing adipocytes.72 Quite a few research have demonstrated that diosgenin can regulate blood lipids by eradicating extreme free radicals and lowering physiological lipid deposition.
Regulating Lipase Exercise
Hepatic lipoprotein lipase (LPL) and hepatic lipase (HL) are essential within the technique of lipoprotein hydrolysis. As soon as lipoproteins are transported to the circulation, particular lipases may also help to launch their lipid elements. The secretory dimer lipases, LPL and HL, hydrolyze triglycerides in lipoproteins and supply FFA for tissues. LPL is secreted from the parenchyma of adipose and muscle to the capillary lumen and acts on VLDL and chylous particles. HL is secreted by hepatocytes and acts on residual lipoprotein and HDL wealthy in ldl cholesterol.73 Diosgenin will increase LPL and HL exercise in a dose-dependent method. In contrast with an HFD rat mannequin group, rats handled with diosgenin (132.8 mg/kg) had considerably elevated actions of LPL and HL.74
Pancreatic lipase (PL) is a dietary lipid digestive enzyme, inhibition of which reduces triglyceride digestion and lipid absorption.75 Kwon et al76 demonstrated that dioscin and diosgenin have robust inhibitory results on PL exercise. After remedy with dioscin (100 mg/kg) or diosgenin (100 mg/kg), the elevation of plasma triacylglycerol focus was considerably lowered in mice orally injected with corn oil. Additional, in an 8-week experiment, rats fed with a HFD containing 2% or 5% Dioscorea nipponica powder exhibited considerably suppressed weight achieve and prevented dietary fats absorption relative to controls fed the HFD alone. The cholesterol-lowering impact of saponins and sapogenins has been broadly described in literature concerning the remedy of hyperlipidemia. Navarro Del Hierro et al77 discovered that saponin-rich extracts and their hydrolysates from fenugreek and quinoa can inhibit PL exercise. Though the precise function of sapogenins contained within the hydrolyzed extracts in lipase inhibition stays unclear, the research offered a possible baseline for the event of multi-bioactive merchandise that act towards pancreatic lipase and ldl cholesterol absorption concurrently. In abstract, diosgenin lowered dietary fats decomposition and absorption within the digestive organs by lowering the hydrolytic exercise of those key enzymes, thus assuaging the signs of metabolic illnesses, akin to hyperlipidemia. In future, the regulation of lipase exercise by diosgenin warrants additional analysis.
Regulating Transcription Components Associated to Lipid Metabolism
The metabolic enzymes and receptors concerned in lipid metabolism are regulated by varied transcription components. Molecules established as concerned in lipid metabolism embody SREBPs,78,79 LXRs,80,81 and peroxisome proliferator activated receptors (PPAR).82,83
SREBPs instantly activate the expression of greater than 30 genes, which contribute to the synthesis and uptake of ldl cholesterol, fatty acids, triglycerides, and phospholipids, in addition to nicotinamide adenine dinucleotide phosphate cofactors required for the synthesis of those molecules.84 SREBP-1c preferentially enhances the transcription of genes required for fatty acid synthesis, which is a key regulator of lipid synthesis and accumulation.85 Diosgenin can inhibit the elevated expression of the sterol regulatory element-binding protein, SREBP-1, and that of its goal genes, together with FAS, SCD-1, and ACC.86 Mechanistic research demonstrated that dioscin can regulate the expression ranges of downstream proteins, together with SREBP-1c, carnitine palmitoyl transferase, FAS, SCD, forkhead field O1, and adipose triglyceride lipase, by regulating the Silent data regulator of transcription 1 (SIRT1)/AMPK signaling pathway, thus considerably lowering lipid metabolism.87 One research reported vital down-regulation of ACC and FAS, alongside up-regulation of PPARγ and LDL receptor (LDLR), genes on the mRNA stage in SW480 cell traces handled with 6.21 µg/mL diosgenin.88 These findings point out that SREBP is a key hub in diosgenin regulation of irregular lipid metabolism.
LXRs, notably liver LXRα, have essential roles within the transcriptional regulation of lipid metabolism. As a ldl cholesterol sensor, the nuclear receptor, LXR, is a vital consider regulating ldl cholesterol homeostasis. LXRα activation can promote the RCT course of, ldl cholesterol catabolism, and ldl cholesterol excretion.81 Against this, LXR agonists enhance hepatic and plasma ranges of triglyceride in mice, owing to enhanced fatty acid biosynthesis and VLDL secretion.89 Activated LXRs can promote ldl cholesterol metabolism; nevertheless, they inevitably produce classical lipogenic unwanted effects. SREBP1 is a major goal of LXRα in upregulating lipogenesis, LXR enhances fatty acid synthesis by activating SREBP1c transcription, which in flip transactivates lipogenic genes. LXR additionally instantly stimulates the transcription of particular lipogenic genes, together with acetyl-CoA carboxylase α (ACACA) in chick embryos and fatty acid synthase (FASN) in human cells.90 Diosgenin (10, 25, and 50 µM) considerably inhibited the buildup of TG and the rise of SREBP-1c mRNA in HepG2 cells induced by excessive glucose ranges. As well as, in contrast with the mannequin group, diosgenin (0.5% or 1% w/w) handled rats fed with a HFD additionally exhibited considerably suppressed LXRα ranges.91 These information exhibit that diosgenin could affect fatty acid metabolism by regulating the LXRα/SREBP-1c signaling pathway. Up to now, there was inadequate research of whether or not diosgenin can regulate ldl cholesterol metabolism-related pathways via LXRs to keep up ldl cholesterol homeostasis. It will likely be of nice significance to additional confirm the precise mechanism underlying diosgenin-mediated regulation of LXRs.
PPARs are members of the nuclear receptor superfamily, and their physiological capabilities are associated to metabolism, vitality homeostasis, and cell improvement and differentiation. Three PPARs have been recognized: PPARα, PPARγ, and PPARβ/δ.92 Many PPAR agonists have been synthesized to deal with metabolic illnesses, notably dyslipidemia and sort 2 diabetes mellitus (T2DM), attributable to their essential metabolic regulatory roles and wonderful druggability.93 Hepatic PPARα stimulates fatty acid catabolism by modulating the expression of LPL, apolipoprotein genes, fatty acid transport and oxidation genes, and genes concerned in HDL metabolism and ketone synthesis. Subsequently, PPARα activators have very important roles dyslipidemia remedy.94 Sangeetha et al68 detected a slight enhance in PPARα expression in 3T3-L1 cells handled with diosgenin at 0.5, 1, and 10 µM, however the enhance was not as vital as that of PPARγ. PPARγ is especially expressed in adipose tissue, hematopoietic cells, and the massive gut, and has key roles in lipid and glucose metabolism.95 PPARγ agonists are broadly used for T2DM remedy and their scientific efficacy as oral antidiabetic brokers is effectively established;96 nevertheless, there’s controversy in regards to the impact of diosgenin on PPARγ, one research confirmed that diosgenin can down-regulate PPARγ expression and exercise, and inhibit adipocyte differentiation.97 One other research confirmed that diosgenin is a selective agonist of PPARγ and up-regulates its expression. This can be why adipogenesis was noticed in 3T3-L1 cells, leading to lowered circulating free lipids in diabetic animals, and diosgenin could also be thought-about as a hypolipidemic drug.68 At current, the mechanism underlying of the results of diosgenin on PPARs are unclear. In silico docking research confirmed that PPARα and PPARγ each work together with diosgenin, whereas the effectivity of diosgenin binding to PPARγ is greater than that of PPARα.68 This raises a query of whether or not diosgenin could also be a twin agonist of each PPARα and PPARγ. A latest research confirmed that integrative utility of PPARα and PPARγ agonists could lower hepatic lipid accumulation, oxidative stress, and manufacturing of inflammatory cytokines, which can be as a result of synergistic impact of PPARα and PPARγ in regulating the expression of the downstream goal genes, SREBP-1c, FAS, DGAT, LPL, and NF-jB.98 As artificial ligands have many unwanted effects,99 utilizing pure agonists, akin to diosgenin, could also be a protected various technique for focused regulation of lipid metabolism. Diosgenin will also be thought-about a twin agonist, which may present a greater steadiness between efficacy and unwanted effects, in contrast with single agonists or twin agonists with various efficiency.
In abstract, diosgenin participates in regulating lipid metabolism via a number of targets and pathways. The underlying mechanisms are extremely complicated and extra animal research and scientific trials are wanted to unravel the main points sooner or later.
Benefits and Limitations of Diosgenin
Plant extracts and their derivatives are broadly used to deal with varied illnesses due to their excessive bioavailability, low unwanted effects, and low value. Lately, diosgenin and its derivatives have been the main focus of appreciable analysis efforts worldwide. Quite a few research have elucidated the pharmacological results of diosgenin and its derivatives on varied illnesses, akin to most cancers,23 diabetes,100 and osteoporosis.101 Diosgenin will be obtained from varied medicinal crops, notably fenugreek seeds, which symbolize a complete and low-cost supply. Generally, diosgenin and different plant-derived compounds have decrease toxicity than chemical medication. Notably, diosgenin has good cytotoxic exercise towards HepG2 cells, however comparatively low toxicity to L02 cells, indicating a level of selectivity between regular and tumor cells.102 We infer that steroidal saponins, together with diosgenin, don’t present any vital toxicity at typical dosages. Because of the poor water solubility and powerful hydrophobicity of diosgenin, its oral bioavailability isn’t splendid, and quite a few diosgenin formulation methods have been utilized to beat these points, together with liquid crystalline (LCs) and b-cyclodextrin (b-CD) complexes, nanocrystals, and soluplus-mediated diosgenin Amorphous Strong Dispersion (ASD) expertise.103,104 In latest many years, ASDs have been the topic of elevated analysis curiosity, as they’ll enhance the oral bioavailability of poorly soluble medication.105 The aqueous solubility of optimized ASDs was considerably improved as a result of amorphization of diosgenin and the molecular interactions between diosgenin and soluplus. Moreover, pharmacokinetic research in rats revealed that the bioavailability of diosgenin from ASDs was improved roughly five-fold. Therefore, diosgenin ASDs, with their excessive solubility, excessive bioavailability, and excessive stability, symbolize a promising route towards pharmaceutical functions.104 Concurrently, though quite a few preclinical research have demonstrated that diosgenin has dependable lipid metabolism regulation operate, scientific analysis information stay restricted. Subsequently, it is going to be essential to confirm the therapeutic results of diosgenin on human hyperlipidemia by way of extra scientific trials sooner or later.
Conclusion and Future Prospects
Phytochemicals present in medicinal or edible crops have exceptional organic exercise, excessive security, low or no toxicity, and symbolize various sources for creating new medication. Diosgenin is a pure steroidal saponin, with potential for broad utility. Quite a few research have confirmed the nice potential to be used of diosgenin within the remedy and prevention of hyperlipidemia. On this paper, we reviewed the pharmacological mechanism underlying the results of diosgenin within the remedy of hyperlipidemia, together with its roles in inhibition of intestinal lipid absorption, regulation of ldl cholesterol transport, promotion of ldl cholesterol conversion into bile acid and excretion, inhibition of endogenous lipid biosynthesis, results on antioxidation and lipoprotein lipase exercise, and regulation of transcription components associated to lipid metabolism. Mixed with analysis progress concerning the pathological mechanism underlying dyslipidemia, associated pathways have been mentioned. Medicine to deal with dyslipidemia primarily scale back blood lipids by regulating the kinetics of various elements of the metabolic cycle. Researchers worldwide are utilizing community pharmacology,106 chromatography, mass spectrometry,107 and GCMS108 to check the pathogenesis and remedy of hyperlipidemia and the ensuing findings will present a scientific foundation for understanding the “multi component-multi target-multi pathway” mechanisms underlying the results of diosgenin on hyperlipidemia.
As well as, a number of new diosgenin supply programs have been developed by combining diosgenin with b-CD, LC, nanocrystalline, and ADS applied sciences, to enhance the water solubility and bioavailability of the compound, which has been helpful to the scientific utility and organic exercise of diosgenin. Medicine containing diosgenin are already marketed in China; nevertheless, there have been few studies on scientific trials of diosgenin for treating dyslipidemia, making it’s tough to evaluate the potential scientific advantages of diosgenin for treating human hyperlipidemia. Thus, extra scientific trials are wanted to make sure the effectiveness and security of diosgenin when used to control human lipid metabolism. Diosgenin may very well be used as a small molecule probe to seek for natural-binding companions in vivo that play essential roles in lowering blood lipids. Sooner or later, with additional research on the mechanisms underlying diosgenin exercise, diosgenin will be anticipated to play a safer and simpler pharmacological function and be appropriately utilized within the remedy of illness within the clinic.
Abbreviations
CHD, coronary coronary heart illness; VLDL-C, very low-density lipoprotein ldl cholesterol; LDL-C, low-density lipoprotein ldl cholesterol; HDL-C, high-density lipoprotein ldl cholesterol; CVD, heart problems; CV, cardiovascular; VLDL, very low-density lipoprotein; LDL, low-density lipoprotein; LDLR, low-density lipoprotein receptor; IDL, medium-density lipoprotein; HDL, high-density lipoprotein; SD, Sprague–Dawley; HFD, high-fat food plan; HCD, high-cholesterol food plan; NPC1L1, Niemann-Choose C1-Like 1; SREBPs, Sterol regulatory element-binding proteins; SREBP-1, sterol regulatory element-binding protein; SREBP-2, sterol regulatory element-binding protein-2; MPD, methyl protodioscin; ABCA1, ATP binding cassette A1; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; HMGCR, 3-hydroxy-3-methylglutaryl-coenzyme A reductase; FAS, fatty acid synthase; ACC, acyl-CoA carboxylase; SRB1, scavenger receptor class B sort I; RCT, reverse transport of ldl cholesterol; CE, ldl cholesterol ester; CES-1, carboxylesterase 1; FC, free ldl cholesterol; CYP7A1, ldl cholesterol 7α-hydroxylase; BA, bile acid; FXR, farnesol X receptor; FAS, fatty acid synthase; SCD-1, stearoyl-CoA desaturase 1; ACC, acetyl-CoA carboxylase; HepG2, hepatocellular carcinoma; AMPK, adenosine 5‘-monophosphate-activated protein kinase; FFA, free fatty acids; LXR, liver X receptor; LXRα, liver X receptor alpha; ABCG5, ATP-binding cassette transporters G5; ABCG5, ATP-binding cassette transporters G8; PL, phospholipids; ApoA-I, apolipoprotein A-I; PPD, pseudoprotodioscin; SOD, superoxide dismutase; GPx, glutathione peroxidase; GSH, glutathione; TBARS, thiobarbituric acid reactive substances; ROS, reactive oxygen species; STZ, streptozotocin–Induced; CRF, power renal failure; eNOS, endothelial nitric oxide synthase; ACE, angiotensin changing enzyme; MDA, malondialdehyde; LPL, lipoprotein lipase; HL, hepatic lipase; PL, pancreatic lipase; PPAR, peroxisome proliferator activated receptors; NADPH, nicotinamide adenine dinucleotide phosphate; SIRT1, silent data regulator of transcription 1; ACACA, acetyl-CoA carboxylase α; FASN, fatty acid synthase; T2DM, sort 2 diabetes mellitus; L02, regular human liver cells; LCs, liquid crystalline; b-CD, b-cyclodextrin; ASD, amorphous stable dispersion; SPF, specific-pathogen-free; Mest, mesoderm particular transcript; MCP-1, monocyte chemoattractant protein; CM, chylomicron; PCSK9, proprotein convertase subtilisin/kexin sort 9.
Acknowledgments
We wish to thank Liping Zhang for her well timed assist in revising the grammar and logic of the manuscript. This paper was supported by the Nationwide Pure Science Basis of China (No. 81573945), the Science and Know-how Growth Venture of Shandong Province (No. 2013GSF11902), the Nationwide Prestigious Chinese language Drugs Physician Studio of Xinlu Wang Venture ([2016]47), and the Science and Know-how Growth Venture of Conventional Chinese language Drugs in Shandong Province (Nos. 2013-081 and 2019-0093).
Disclosure
The authors report no conflicts of curiosity on this work.
References
1. Karr S. Epidemiology and administration of hyperlipidemia. Am J Manag Care. 2017;23(9 Suppl):S139–S148.
2. Benjamin EJ, Virani SS, Callaway CW, et al. Coronary heart illness and stroke statistics-2018 replace: a report from the American coronary heart affiliation. Circulation. 2018;137(12):e67–e492.
3. Townsend N, Nichols M, Scarborough P, Rayner M. Heart problems in Europe–epidemiological replace 2015. Eur Coronary heart J. 2015;36(40):2696–2705. doi:10.1093/eurheartj/ehv428
4. Wang XG, Zhao X, Li Y, et al. Analysis progress of pathogenesis and remedy of hyperlipidemia. J Liaoning Univ Trad Chin Med. 2020;22(12):196–200.
5. Kopin L, Lowenstein C. Dyslipidemia. Ann Intern Med. 2017;167(11):ITC81–ITC96. doi:10.7326/AITC201712050
6. Holven KB, Ulven SM, Bogsrud MP. Hyperlipidemia and heart problems with give attention to familial hypercholesterolemia. Curr Opin Lipidol. 2017;28(5):445–447. doi:10.1097/MOL.0000000000000449
7. Strilchuk L, Fogacci F, Cicero AF. Security and tolerability of injectable lipid-lowering medication: an replace of scientific information. Knowledgeable Opin Drug Saf. 2019;18(7):611–621. doi:10.1080/14740338.2019.1620730
8. Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle signs: affect on statin therapy-European Atherosclerosis Society consensus panel assertion on evaluation, aetiology and administration. Eur Coronary heart J. 2015;36(17):1012–1022. doi:10.1093/eurheartj/ehv043
9. Mach F, Baigent C, Catapano AL, et al. 2019 ESC/EAS pointers for the administration of dyslipidaemias: lipid modification to cut back cardiovascular danger. Eur Coronary heart J. 2020;41(1):111–188.
10. Thomford NE, Senthebane DA, Rowe A, et al. Pure merchandise for drug discovery within the twenty first century: improvements for novel drug discovery. Int J Mol Sci. 2018;19(6):1578. doi:10.3390/ijms19061578
11. Talebi S, Bagherniya M, Atkin SL, Askari G, Orafai HM, Sahebkar A. The helpful results of nutraceuticals and pure merchandise on small dense LDL ranges, LDL particle quantity and LDL particle measurement: a scientific assessment. Lipids Well being Dis. 2020;19(1):66. doi:10.1186/s12944-020-01250-6
12. Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, et al. Discovery and resupply of pharmacologically lively plant-derived pure merchandise: a assessment. Biotechnol Adv. 2015;33(8):1582–1614.
13. Rodrigues T, Reker D, Schneider P, Schneider G. Relying on pure merchandise for drug design. Nat Chem. 2016;8(6):531–541. doi:10.1038/nchem.2479
14. Chen Y, Tang YM, Yu SL, et al. Advances within the pharmacological actions and mechanisms of diosgenin. Chin J Nat Med. 2015;13(8):578–587.
15. Yuan B, Byrnes DR, Dinssa FF, Simon JE, Wu Q. Identification of polyphenols, glycoalkaloids, and saponins in Solanum scabrum berries utilizing HPLC-UV/Vis-MS. J Meals Sci. 2019;84(2):235–243. doi:10.1111/1750-3841.14424
16. Krol-Kogus B, Lamine KM, Migas P, Boudjeniba M, Krauze-Baranowska M. HPTLC dedication of diosgenin in fenugreek seeds. Acta Pharm. 2018;68(1):97–107. doi:10.2478/acph-2018-0002
17. Gong J, Fang Okay, Dong H, Wang D, Hu M, Lu F. Impact of fenugreek on hyperglycaemia and hyperlipidemia in diabetes and prediabetes: a meta-analysis. J Ethnopharmacol. 2016;194:260–268. doi:10.1016/j.jep.2016.08.003
18. Wu FC, Jiang JG. Results of diosgenin and its derivatives on atherosclerosis. Meals Funct. 2019;10(11):7022–7036. doi:10.1039/C9FO00749K
19. Cai B, Zhang Y, Wang Z, et al. Therapeutic potential of diosgenin and its main derivatives towards neurological illnesses: latest advances. Oxid Med Cell Longev. 2020;2020:3153082. doi:10.1155/2020/3153082
20. Wu S, Zhao F, Zhao J, et al. Dioscin improves postmenopausal osteoporosis via inducing bone formation and inhibiting apoptosis in ovariectomized rats. Biosci Tendencies. 2019;13(5):394–401. doi:10.5582/bst.2019.01186
21. Li R, Liu Y, Shi J, et al. Diosgenin regulates ldl cholesterol metabolism in hypercholesterolemic rats by inhibiting NPC1L1 and enhancing ABCG5 and ABCG8. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(8):1124–1133. doi:10.1016/j.bbalip.2019.04.010
22. Tao X, Yin L, Xu L, Peng J. Dioscin: a various performing pure compound with therapeutic potential in metabolic illnesses, most cancers, irritation and infections. Pharmacol Res. 2018;137:259–269. doi:10.1016/j.phrs.2018.09.022
23. Sethi G, Shanmugam MK, Warrier S, et al. Professional-apoptotic and anti-cancer properties of diosgenin: a complete and significant assessment. Vitamins. 2018;10(5):5. doi:10.3390/nu10050645
24. Cayen MN, Dvornik D. Impact of diosgenin on lipid metabolism in rats. J Lipid Res. 1979;20(2):162–174. doi:10.1016/S0022-2275(20)40628-5
25. Liu MX, Zhang FX, Xun LY, Guan DH. Research on regulating blood lipid results and the mechanism of diosgenin. Arch Tradit Chin Med. 2016;34(04):798–800.
26. Parama D, Boruah M, Kumari Y, et al. Diosgenin, a steroidal saponin, and its analogs: efficient therapies towards completely different power illnesses. Life Sci. 2020;260:118182.
27. Liao AM, Jung H, Yu JW, et al. Synthesis and organic analysis of arginyl-diosgenin conjugate as a possible bone tissue engineering agent. Chem Biol Drug Des. 2018;91(1):17–28. doi:10.1111/cbdd.13050
28. Chen FX, Zhao MR, Ren BZ, et al. Solubility of diosgenin in numerous solvents. J Chem Thermodyn. 2012;47:341–346. doi:10.1016/j.jct.2011.11.009
29. Chaudhary SA, Chaudhary PS, Syed BA, et al. Validation of a way for diosgenin extraction from fenugreek (Trigonella foenum-graecum L.). Acta Sci Pol Technol Aliment. 2018;17(4):377–385.
30. Selim S, Al Jaouni S. Anticancer and apoptotic results on cell proliferation of diosgenin remoted from Costus speciosus (Koen.) Sm. BMC Complement Altern Med. 2015;15(1):301. doi:10.1186/s12906-015-0836-8
31. Kim JK, Park SU. An replace on the organic and pharmacological actions of diosgenin. EXCLI J. 2018;17:24–28.
32. Li X, Wang Y, Solar J, et al. Chemotaxonomic research of 12 Dioscorea species from China by UHPLC-QTOF-MS/MS evaluation. Phytochem Anal. 2020;31(2):164–182. doi:10.1002/pca.2876
33. Herrera T, Navarro Del Hierro J, Fornari T, Reglero G, Martin D. Acid hydrolysis of saponin-rich extracts of quinoa, lentil, fenugreek and soybean to yield sapogenin-rich extracts and different bioactive compounds. J Sci Meals Agric. 2019;99(6):3157–3167. doi:10.1002/jsfa.9531
34. Sahu P, Gidwani B, Dhongade HJ. Pharmacological actions of dehydroepiandrosterone: a assessment. Steroids. 2020;153:108507. doi:10.1016/j.steroids.2019.108507
35. Dong J, Lei C, Lu D, Wang Y. Direct biotransformation of dioscin into diosgenin in rhizome of Dioscorea zingiberensis by penicillium dioscin. Indian J Microbiol. 2015;55(2):200–206. doi:10.1007/s12088-014-0507-3
36. Sauvaire Y, Ribes G, Baccou JC, Loubatieères-Mariani MM. Implication of steroid saponins and sapogenins within the hypocholesterolemic impact of fenugreek. Lipids. 1991;26(3):191–197. doi:10.1007/BF02543970
37. Wang T, Choi RC, Li J, et al. Antihyperlipidemic impact of protodioscin, an lively ingredient remoted from the rhizomes of Dioscorea nipponica. Planta Med. 2010;76(15):1642–1646. doi:10.1055/s-0030-1249960
38. Malinow MR, Elliott WH, McLaughlin P, Upson B. Results of artificial glycosides on steroid steadiness in Macaca fascicularis. J Lipid Res. 1987;28(1):1–9. doi:10.1016/S0022-2275(20)38729-0
39. Yamanashi Y, Takada T, Yamamoto H, Suzuki H. NPC1L1 facilitates sphingomyelin absorption and regulates diet-induced manufacturing of VLDL/LDL-associated S1P. Vitamins. 2020;12(9):2641. doi:10.3390/nu12092641
40. Tang W, Lin J, Ma Y, et al. Ezetimibe restores biliary ldl cholesterol excretion in mice expressing Niemann–Choose C1-like 1 solely in liver. Biochim Biophys Acta. 2011;1811(9):549–555. doi:10.1016/j.bbalip.2011.05.013
41. Ma HY, Zhao ZT, Wang LJ, Wang Y, Zhou QL, Wang BX. Comparative research on anti-hypercholesterolemia exercise of diosgenin and complete saponin of Dioscorea panthaica. J Trad Chin Med. 2002;27(7):51–54.
42. Kuang SY, Li Y, Kuang WD. Results of diosgenin on ldl cholesterol metabolism and caveolin-1 expression in human hepatocyte L-02. Chin Pharm. 2013;24(35):3286–3288.
43. Rhee EJ, Byrne CD, Sung KC. The HDL ldl cholesterol/apolipoprotein A-I ratio: an indicator of heart problems. Curr Opin Endocrinol Diabetes Obes. 2017;24(2):148–153. doi:10.1097/MED.0000000000000315
44. Kusano Y, Tsujihara N, Masui H, Kozai H, Takeuchi W. Consumption of Japanese yam improves lipid metabolism in high-cholesterol diet-fed rats. Nutr Sci Vitaminol (Tokyo). 2016;62(5):350–360. doi:10.3177/jnsv.62.350
45. Ma W, Ding H, Gong X, et al. Methyl protodioscin will increase ABCA1 expression and ldl cholesterol efflux whereas inhibiting gene expressions for synthesis of ldl cholesterol and triglycerides by suppressing SREBP transcription and microRNA 33a/b ranges. Atherosclerosis. 2015;239(2):566–570. doi:10.1016/j.atherosclerosis.2015.02.034
46. Shen WJ, Azhar S, Kraemer FB. SR-B1: a singular multifunctional receptor for ldl cholesterol inflow and efflux. Annu Rev Physiol. 2018;80:95–116. doi:10.1146/annurev-physiol-021317-121550
47. Xu J, Xu Y, Xu Y, Yin L, Zhang Y. International inactivation of carboxylesterase 1 (Ces1/Ces1g) protects towards atherosclerosis in Ldlr−/− mice. Sci Rep. 2017;7(1):17845. doi:10.1038/s41598-017-18232-x
48. Jones H, Alpini G, Francis H. Bile acid signaling and biliary capabilities. Acta Pharm Sin B. 2015;5(2):123–128. doi:10.1016/j.apsb.2015.01.009
49. Yu L, Lu H, Yang X, et al. Diosgenin alleviates hypercholesterolemia by way of SRB1/CES-1/CYP7A1/FXR pathway in high-fat diet-fed rats. Toxicol Appl Pharmacol. 2021;412:115388. doi:10.1016/j.taap.2020.115388
50. Gesto DS, Pereira C, Cerqueira NM, Sousa SF. An atomic-level perspective of HMG-CoA-reductase: the goal enzyme to deal with hypercholesterolemia. Molecules. 2020;25(17):3891. doi:10.3390/molecules25173891
51. Fox KM, Tai M-H, Kostev Okay, Hatz M, Qian Y, Laufs U. Remedy patterns and low-density lipoprotein ldl cholesterol (LDL-C) purpose attainment amongst sufferers receiving high- or moderate-intensity statins. Clin Res Cardiol. 2017;107(5):380–388. doi:10.1007/s00392-017-1193-z
52. Hao S, Xu R, Li D, Zhu Z, Wang T, Liu Okay. Attenuation of streptozotocin-induced lipid profile anomalies within the coronary heart, mind, and mRNA expression of HMG-CoA reductase by diosgenin in rats. Cell Biochem Biophys. 2015;72(3):741–749. doi:10.1007/s12013-015-0525-8
53. Uemura T, Goto T, Kang MS, et al. Diosgenin, the principle aglycon of fenugreek, inhibits LXRalpha exercise in HepG2 cells and reduces plasma and hepatic triglycerides in overweight diabetic mice. J Nutr. 2011;141(1):17–23. doi:10.3945/jn.110.125591
54. Pan CH, Tsai CH, Liu FC, et al. Affect of various particle processing on hypocholesterolemic and antiatherogenic actions of yam (Dioscorea pseudojaponica) in cholesterol-fed rabbit mannequin. Sci Meals Agric. 2013;93(6):1278–1283. doi:10.1002/jsfa.5882
55. Carling D. AMPK signalling in well being and illness. Curr Opin Cell Biol. 2017;45:31–37. doi:10.1016/j.ceb.2017.01.005
56. Cheng SL. The Motion and Mechanism of Diosgenin Suppress the Nonalcoholic Fatty Liver Illness. Second Army Medical College; 2014.
57. Poudel B, Lim SW, Ki HH, Nepali S, Lee YM, Kim DK. Dioscin inhibits adipogenesis via the AMPK/MAPK pathway in 3T3-L1 cells and modulates fats accumulation in overweight mice. Int J Mol Med. 2014;34(5):1401–1408. doi:10.3892/ijmm.2014.1921
58. Ying F, Cai Y, Wong HK, et al. EP4 emerges as a novel regulator of bile acid synthesis and its activation protects towards hypercholesterolemia. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(9):1029–1040. doi:10.1016/j.bbalip.2018.06.003
59. Liu S, Jing F, Yu C, Gao L, Qin Y, Zhao J. AICAR-induced activation of AMPK inhibits TSH/SREBP-2/HMGCR pathway in liver. PLoS One. 2015;10(5):e0124951. doi:10.1371/journal.pone.0124951
60. Patel SB, Graf GA, Temel RE. ABCG5 and ABCG8: greater than a protection towards xenosterols. J Lipid Res. 2018;59(7):1103–1113. doi:10.1194/jlr.R084244
61. Temel RE, Brown JM, Ma Y, et al. Diosgenin stimulation of fecal ldl cholesterol excretion in mice isn’t NPC1L1 dependent. J Lipid Res. 2009;50(5):915–923. doi:10.1194/jlr.M800631-JLR200
62. Phillips MC. Is ABCA1 a lipid switch protein? J Lipid Res. 2018. https://pubmed.ncbi.nlm.nih.gov/29305383/ references
63. Lv YC, Yang J, Yao F, et al. Diosgenin inhibits atherosclerosis by way of suppressing the MiR-19b-induced downregulation of ATP-binding cassette transporter A1. Atherosclerosis. 2015;240(1):80–89. doi:10.1016/j.atherosclerosis.2015.02.044
64. Gai Y, Li Y, Xu Z, Chen J. Pseudoprotodioscin inhibits SREBPs and microRNA 33a/b ranges and reduces the gene expression concerning the synthesis of ldl cholesterol and triglycerides. Fitoterapia. 2019;139:104393. doi:10.1016/j.fitote.2019.104393
65. Marrocco I, Altieri F, Peluso I. Measurement and scientific significance of biomarkers of oxidative stress in people. Oxid Med Cell Longev. 2017;2017:6501046. doi:10.1155/2017/6501046
66. Gupta DD, Mishra S, Verma SS, et al. Analysis of antioxidant, anti-inflammatory and anticancer actions of diosgenin enriched Paris polyphylla rhizome extract of Indian Himalayan landraces. J Ethnopharmacol. 2021;270:113842. doi:10.1016/j.jep.2021.113842
67. Pi WX, Feng XP, Ye LH, Cai BC. Mixture of morroniside and diosgenin prevents excessive glucose-induced cardiomyocytes apoptosis. Molecules. 2017;22(1):163. doi:10.3390/molecules22010163
68. Sangeetha MK, ShriShri Mal N, Atmaja Okay, Sali VK, Vasanthi HR. PPAR’s and diosgenin a chemico organic perception in NIDDM. Chem Biol Work together. 2013;206(2):403–410. doi:10.1016/j.cbi.2013.08.014
69. Manivannan J, Balamurugan E, Silambarasan T, Raja B. Diosgenin improves vascular operate by rising aortic eNOS expression, normalize dyslipidemia and ACE exercise in power renal failure rats. Mol Cell Biochem. 2013;384(1–2):113–120. doi:10.1007/s11010-013-1788-2
70. Xu T, Zheng L, Xu L, et al. Protecting results of dioscin towards alcohol-induced liver harm. Arch Toxicol. 2014;88(3):739–753. doi:10.1007/s00204-013-1148-8
71. Son IS, Kim JH, Sohn HY, Son KH, Kim JS, Kwon CS. Antioxidative and hypolipidemic results of diosgenin, a steroidal saponin of yam (Dioscorea spp.), on high-cholesterol fed rats. Biosci Biotechnol Biochem. 2007;71(12):3063–3071. doi:10.1271/bbb.70472
72. Uemura T, Hirai S, Mizoguchi N, et al. Diosgenin current in fenugreek improves glucose metabolism by selling adipocyte differentiation and inhibiting irritation in adipose tissues. Mol Nutr Meals Res. 2010;54(11):1596–1608. doi:10.1002/mnfr.200900609
73. Koerner CM, Roberts BS, Neher SB. Endoplasmic reticulum high quality management in lipoprotein metabolism. Mol Cell Endocrinol. 2019;498:110547. doi:10.1016/j.mce.2019.110547
74. Gong G, Qin Y, Huang W, et al. Protecting results of diosgenin within the hyperlipidemic rat mannequin and in human vascular endothelial cells towards hydrogen peroxide-induced apoptosis. Chem Biol Work together. 2010;184(3):366–375. doi:10.1016/j.cbi.2010.02.005
75. Matsuo Y, Matsumoto Okay, Inaba N, Mimaki Y. Daisaikoto inhibits pancreatic lipase exercise and reduces serum triglyceride ranges in mice. Biol Pharm Bull. 2018;41(9):1485–1488. doi:10.1248/bpb.b18-00324
76. Kwon CS, Sohn HY, Kim SH, et al. Anti-obesity impact of Dioscorea nipponica Makino with lipase-inhibitory exercise in rodents. Biosci Biotechnol Biochem. 2003;67(7):1451–1456. doi:10.1271/bbb.67.1451
77. Navarro Del Hierro J, Casado-Hidalgo G, Reglero G, Martin D. The hydrolysis of saponin-rich extracts from fenugreek and quinoa improves their pancreatic lipase inhibitory exercise and hypocholesterolemic impact. Meals Chem. 2021;338:128113. doi:10.1016/j.foodchem.2020.128113
78. Zhang F, Solar W, Chen J, et al. SREBP-2, a brand new goal of metformin? Drug Des Devel Ther. 2018;12:4163–4170. doi:10.2147/DDDT.S190094
79. Shimano H, Sato R. SREBP-regulated lipid metabolism: convergent physiology – divergent pathophysiology. Nat Rev Endocrinol. 2017;13(12):710–730.
80. Schulman IG. Liver X receptors hyperlink lipid metabolism and irritation. FEBS Lett. 2017;591(19):2978. doi:10.1002/1873-3468.12702
81. Wang B, Tontonoz P. Liver X receptors in lipid signalling and membrane homeostasis. Nat Rev Endocrinol. 2018;14(8):452–463. doi:10.1038/s41574-018-0037-x
82. de la Rosa Rodriguez MA, Kersten S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862(10 Pt B):1212–1220. doi:10.1016/j.bbalip.2017.07.007
83. Kersten S, Stienstra R. The function and regulation of the peroxisome proliferator activated receptor alpha in human liver. Biochimie. 2017;136:75–84. doi:10.1016/j.biochi.2016.12.019
84. Debose-Boyd RA, Ye J. SREBPs in lipid metabolism, insulin signaling, and past. Tendencies Biochem Sci. 2018;43(5):358-368. doi: 10.1016/j.tibs.2018.01.005.
85. Li X, Li Y, Yang W, et al. SREBP-1c overexpression induces triglycerides accumulation via rising lipid synthesis and lowering lipid oxidation and VLDL meeting in bovine hepatocytes. J Steroid Biochem Mol Biol. 2014;143:174–182. doi:10.1016/j.jsbmb.2014.02.009
86. Hua S, Li Y, Su L, Liu X. Diosgenin ameliorates gestational diabetes via inhibition of sterol regulatory element-binding protein-1. Biomed Pharmacother. 2016;84:1460–1465. doi:10.1016/j.biopha.2016.10.049
87. Yao H, Tao X, Xu L, et al. Dioscin alleviates non-alcoholic fatty liver illness via adjusting lipid metabolism by way of SIRT1/AMPK signaling pathway. Pharmacol Res. 2018;131:51–60. doi:10.1016/j.phrs.2018.03.017
88. Mohammad-Sadeghipour M, Mahmoodi M, Noroozi Karimabad M, Mirzaei MR, Hajizadeh MR. Diosgenin and 4-hydroxyisoleucine from fenugreek are regulators of genes concerned in lipid metabolism within the human colorectal most cancers cell line SW480. Cell J. 2021;22(4):514–522.
89. Schultz JR, Tu H, Luk A, et al. Position of LXRs in command of lipogenesis. Genes Dev. 2000;14(22):2831–2838. doi:10.1101/gad.850400
90. Xu HF, Luo J, Zhang XY, Li J, Bionaz M. Activation of liver X receptor promotes fatty acid synthesis in goat mammary epithelial cells by way of modulation of SREBP1 expression. J Dairy Sci. 2019;102(4):3544–3555. doi:10.3168/jds.2018-15538
91. Cheng S, Liang S, Liu Q, et al. Diosgenin prevents high-fat diet-induced rat non-alcoholic fatty liver illness via the AMPK and LXR signaling pathways. Int J Mol Med. 2018;41(2):1089–1095.
92. Cheng HS, Tan WR, Low ZS, Marvalim C, Lee JYH, Tan NS. Exploration and improvement of PPAR modulators in well being and illness: an replace of scientific proof. Int J Mol Sci. 2019;20(20):5055. doi:10.3390/ijms20205055
93. Mirza AZ, Althagafi II, Shamshad H. Position of PPAR receptor in numerous illnesses and their ligands: physiological significance and scientific implications. Eur J Med Chem. 2019;166:502–513. doi:10.1016/j.ejmech.2019.01.067
94. Bougarne N, Weyers B, Desmet SJ, et al. Molecular actions of PPARα in lipid metabolism and irritation. Endocr Rev. 2018;39(5):760–802.
95. Janani C, Ranjitha Kumari BD. PPAR gamma gene–a assessment. Diabetes Metab Syndr. 2015;9(1):46–50. doi:10.1016/j.dsx.2014.09.015
96. Alam F, Islam MA, Mohamed M, et al. Efficacy and security of pioglitazone monotherapy in sort 2 diabetes mellitus: a scientific assessment and meta-analysis of randomised managed trials. Sci Rep. 2019;9(1):5389. doi:10.1038/s41598-019-41854-2
97. Wang X, Liu J, Lengthy Z, et al. Impact of diosgenin on metabolic dysfunction: function of ERbeta within the regulation of PPARgamma. Toxicol Appl Pharmacol. 2015;289(2):286–296. doi:10.1016/j.taap.2015.09.015
98. Zhang Y, Cui Y, Wang XL, Shang X, Xie ML. PPARα/γ agonists and antagonists in a different way have an effect on hepatic lipid metabolism, oxidative stress and inflammatory cytokine manufacturing in steatohepatitic rats. Cytokine. 2015;75(1):127–135. doi:10.1016/j.cyto.2015.05.031
99. Nanjan MJ, Mohammed M, Prashantha Kumar BR, Chandrasekar MJN. Thiazolidinediones as antidiabetic brokers: a crucial assessment. Bioorg Chem. 2018;77:548–567. doi:10.1016/j.bioorg.2018.02.009
100. Gan Q, Wang J, Hu J, et al. The function of diosgenin in diabetes and diabetic issues. J Steroid Biochem Mol Biol. 2020;198:105575. doi:10.1016/j.jsbmb.2019.105575
101. Zhao S, Niu F, Xu CY, et al. Diosgenin prevents bone loss on retinoic acid-induced osteoporosis in rats. Ir J Med Sci. 2016;185(3):581–587. doi:10.1007/s11845-015-1309-2
102. Yin H, Zhang MJ, An RF, Zhou J, Huang XF. Diosgenin derivatives as potential antitumor brokers: synthesis, cytotoxicity, and mechanism of motion. J Nat Prod. 2020;84(3):616–29.
103. Manda VK, Avula B, Ali Z, et al. Characterization of in vitro ADME properties of diosgenin and dioscin from Dioscorea villosa. Planta Med. 2013;79(15):1421–1428. doi:10.1055/s-0033-1336521
104. Liu P, Zhou JY, Chang JH, et al. Soluplus-mediated diosgenin amorphous stable dispersion with excessive solubility and excessive stability: improvement, characterization and oral bioavailability. Drug Des Devel Ther. 2020;14:2959–2975. doi:10.2147/DDDT.S253405
105. Schittny A, Huwyler J, Puchkov M. Mechanisms of elevated bioavailability via amorphous stable dispersions: a assessment. Drug Deliv. 2020;27(1):110–127. doi:10.1080/10717544.2019.1704940
106. Banerjee S, Bhattacharjee P, Kar A, Mukherjee PK. LC-MS/MS evaluation and community pharmacology of Trigonella foenum-graecum – a plant from Ayurveda towards hyperlipidemia and hyperglycemia with mixture synergy. Phytomedicine. 2019;60:152944. doi:10.1016/j.phymed.2019.152944
107. Ren Y, Wang Z, Wu C, et al. Ultrahigh-performance liquid chromatography with tandem mass spectrometry for the dedication of 10 alkaloids in beagle plasma after the oral administration of the three Coptidis rhizoma extracts. J Ethnopharmacol. 2019;239:111896. doi:10.1016/j.jep.2019.111896
108. Sozen E, Yazgan B, Sahin A, Ince U, Ozer NK. Excessive ldl cholesterol diet-induced modifications in oxysterol and scavenger receptor ranges in coronary heart tissue. Oxid Med Cell Longev. 2018;2018:8520746. doi:10.1155/2018/8520746