Int J App Pharm, Vol 14, Issue 6, 2022, 9-17Review Article

BIOAVAILABILITY PROBLEMS OF PHYTOSTEROLS: A SYSTEMATIC REVIEW

JAMAL BASHA DUDEKU LA1,2, KUMAR P. R.1*, RANGANAYAKULU D.3

1Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, 2Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh 474020, 3Department of Pharmacology, Sri Padmavathi School of Pharmacy, Tiruchanoor, Tirupati, Andhra Pradesh 517503
Email: jbdudekula@gwa.amity.edu

Received: 05 Jul 2022, Revised and Accepted: 25 Aug 2022


ABSTRACT

Phytosterols (PS) are biologically active steroidal compounds obtained from plant foods and cholesterol is found in animals. They have a prominent role in reducing the low-density lipoprotein (LDL) cholesterol levels, thus decreasing the risk of many diseases. PSs also have anti-cancer, antioxidant, antiulcer, immunomodulatory, antibacterial, antifungal effects and modulate inflammation by promoting the wound healing and inhibition of platelet aggregation. The most challenging part concerned about phytosterols was bioavailability. Phytosterol’s absorption and the concentration of circulation over the body were lesser in human intestine compared to cholesterol because of its selectivity and return through intestinal transporters. We searched PubMed, Scopus, Embase, Google scholar and major conference proceedings. Sixteen such therapeutically potent plant steroids were studied in this systematic review to assess the bioavailability issues of phytosterols. Swiss ADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness was used for the study.

Keywords: Phytosterols, Absorption, Bioavailability, Steroidal compounds


INTRODUCTION

Phytosterols (PS) are biologically active steroidal compounds obtained from plant foods and cholesterol is found in animals [1, 2]. Beta-sitosterol, campesterol, and stigmasterol are the common phytosterols in plant and animal diet such as edible oils, nuts, cereals and citrus fruits [3, 4]. These steroids have a common cholesterol skeleton with different side chains. Phytosterols eminently regulates plenitude of plant physiological process leading to proper growth of the plant [5]. Phytosterols have several health applications but could not be derived by humans; should be obtained from plants and animals as a part of diet. The biological and therapeutic activity of phytosterols depends on their formulation and solubility nature. Researches provide supporting evidences that PSs and their derivatives have multiple pharmacological properties and human-wellness-promoting abilities. They have a prominent role in reducing the low-density lipoprotein (LDL) cholesterol levels, thus decreasing the risk of many diseases [6-8]. PSs also have anti-cancer, antioxidant, antiulcer, immunomodulatory, antibacterial, antifungal effects and modulate inflammation by promoting the wound healing and inhibition of platelet aggregation [9-11].

Therapeutic potential of phytosterols

PSs as an additive therapy proves to have a good therapeutic potential in non-alcoholic fatty liver disease showing improvement in LDL cholesterol and liver enzymes [12]. They act by reducing intestinal absorption of cholesterol and thus regulate the LDL-cholesterol levels in humans. This mechanism is mediated by certain mechanisms like 1) Reducing the amount of cholesterol for absorption by solubilising it in the intestinal lumen, 2) Modifications in the expression of Niemann-Pick C1-like 1 protein which reduce cholesterol transport and promote its efflux from the enterocytes to the intestinal lumen, 3) Trans intestinal cholesterol excretion (TICE) removes the cholesterol from body [13]. In a preclinical study, male C57BL/6 J mice were administered with 3.1% PS and high-fat diet supplementation for three weeks shown a decrease in very-low density lipoprotein (VLDL) secretion [14]. PSs play a crucial role in neurodegenerative diseases. Phytosterols crosses the blood brain barrier and modifies the pathways related to neurodegeneration in the brain. Campesterol, beta sitosterol and stigmasterol are used as a prognostic biomarker for the neurodegenerative disorders like Alzheimer’s disease [15]. Study performed by Rui et al. had documented that a high-cholesterol diet reduce the cognitive performance in animal models [16]. In this study, rats were fed for 6 mo with a high cholesterol diet and 2% g/100 g PS has shown reduced serum lipid levels, improved cognitive performance, triggered an increase in pyramidal cells number. Plasma and hepatic triglycerides (TG) levels were altered by the treatment with phytosterols in laboratory animals. In a study performed by Rideout et al., Syrian golden hamsters fed with a high-fat diet and 2% w/w PSs for 6 w and their TG in blood plasma were found to be decreased [17].

PSs has the ability of promoting glucose metabolism by activating the AMP-activated kinase (AMPK) or peroxisome proliferator-activated receptors (PPARs) in transcriptional regulation pathways [18]. A limited rise in blood glucose scale after oral administration of high glucose concentration and increased insulin response was observed. Study performed on zucker diabetic fatty rats by giving 5-campestenone (0.6%) as dietary supplement has shown minimal rise in blood glucose scale with improved insulin response. ß-sitosterol (20 µM) administration also leads to rise in glucose intake by GLUT4 translocation to the plasma membrane [19]. A study in Db/Db mice fed with 0.3% 5-campestenone for eight weeks has documented a sharp decline in the blood glucose levels and inhibition of glucose elimination [20].

Phytosterols have anticancer properties which stimulates apoptosis by interaction with cell targets. A preclinical study on rats shows that beta sitosterol isolated from Asclepias curassavica L. has dose related effectiveness in colon rectal cancer [21]. Researches proved ß-sitosterol supplementation cause apoptosis by leading to an increase in FAS protein expression, caspase 8 and tumor necrosis factor in MCF-7 and MDA-MB-231 breast cancer cells [22]. ß-sitosterol and daucosterol from Grewia tiliaefolia have the ability to promote apoptosis in A549 lung cancer cells by arresting the cell cycle at the G2/M phase [23]. ß-sitosterol interferes with the DNA fragmentation in the cervical cancer cells in a dose dependent manner.

Phytosterols being similar in structure competes with cholesterol for absorption in the intestine and reduces the LDL cholesterol absorption leading to lipid lowering property [24]. Many formulations with suitable drug delivery vehicles are analysed for oral delivery of phytosterols without affecting its therapeutic efficacy. Formulations of phytosterols incorporated in lecithin micelles, water-dispersible phytosterols with fatty acids and polysorbate composition were able to reduce the serum LDL-cholesterol more efficiently [25, 26]. Nano emulsions, nanoparticles, micro particles, microcapsules, and micelles are some of the formulation approaches used for phytosterols delivery [27]. However, besides increased therapeutic efficacy, the safety and tolerability of these formulations are concerned to phytosterols oxidation products. The prevention of phytosterols oxidation products in the formulation is the major challenge. The studies regarding the ability to reduce phytosterol oxidation products are insufficient [28].

Bioavailability of phytosterols

The most challenging part concerned about phytosterols was bioavailability. Phytosterols absorption and the concentration of circulation over the body were lesser in human intestine compared to cholesterol because of its selectivity and return through intestinal transporters [29]. The absorption of the phytosterols are influenced by several factors like solubility, chemical structure, type of phytosterol as steryl glycoside, sterol or stanol, hydrogenation process, preparation and genetic factors. Despite its studies on wide variety of interesting pharmacologic effects the bioavailability of phytosterols remain as a limiting aspect [30]. The presence of polycyclic nucleus, hydroxyl group at C-3 and C-17 side chain in the structure of phytosterols. The physical and chemical modification of phytosterols enhances the bioavailability. A biologically active substance could be customized into a nano scale delivery system in order to reach its target site. Chemical modification of PSs could be done by esterification and physical modification by microencapsulation. Low solubility and high melting point affects the bioavailability. Physical changes by encapsulation of phytosterols in nanodelivery systems favours dissolution in gastrointestinal tract and improve the bioavailability [31].

Systematic review of phytosterols selected for the study

16 phytosterols were selected in this study and the structures were obtained using chimera software table 1. The drug-likeness parameters like molecular weight and size, melting and boiling points, hydrogen bonding will determine the pharmacological behaviour a compound in aspects of its bioavailability, toxicity, metabolic activity, etc. Molinspiration cheminformatics tool is employed to evaluate the drug likeness properties of the selected phytosterol compounds [32]. The “drug likeness” is defined as the balance between molecular and structural properties of a chemical compound that determines whether that compound is considered as a “drug” [33]. Log P value is calculated for all the 16 selected compounds. The positive log P value indicates that the lipophilic nature (need to cross BBB). Most of the compounds exhibited high logP values indicating better lipophilic nature; except digitonin has negative value and 5 other compounds have log P values less than 5. The water solubility values of all the compounds were found to be negative; thus indicating their low hydrophilic nature table 2.

Swiss ADME software is employed in order to understand the pharmacokinetic and toxicity parameters of a lead compound in the initial drug discovery process itself so as to avoid hindrances in the later stages [34]. The ADME/Toxicity properties depend on the physicochemical parameters namely lipophilicity (logP), molecular weight, polar surface area, molar refractivity and water solubility. The intestinal absorption of orally active drug like compound is mostly assessed by Caco-2 cell model and MDCK (Madin Darby canine kidney) cell. The results of Caco-2 model were almost compatible with the standard human intestinal absorption values. The blood brain barrier penetration of a compound explains its therapeutic ability in the central nervous system; whereas, drug efficacy and disposition is well explained by the in vitro plasma protein binding model. Most of the compounds have their plasma protein binding values low; thus indicating their free distribution nature. Formulation and ease of handling of a chemical compound depends on its water solubility. Oral absorption of a drug is proportional to its water solubility; while, a parenteral drug must be highly water soluble to deliver its active constituents in small volumes. All the PSs shows negative values and hence moderately water soluble table 2.

Table 1: Physical properties

Plant steroids Structure Molecular formula Molecular weight Synonym Log p Boiling point Melting point Phase of the study Therapeutic uses
Campesterol [1] C28H48O 400.7

Campesterin, Campest-5-en-3beta-ol,

Ergost-5-en-3-beta-ol

9.972 489.00 to 490.00 °C 157.5 °C Observational studies [2, 3] Diagnosis of sitosterolemia [4], for the prevention of cancers and cardio metabolic diseases [5], cholesterol absorption biomarker [6]
Ergosterol [7] C28H44O 396.6 Provitamin D2, Ergosterin, ergosta-5,7,22-trien-3-ol 8.86 250 °C at 0.01 mmHg 170 °C Biological precursor of vitamin D2, antirachitic vitamin
Stigmasterol [8] C29H48O 412.7 Beta-stigmasterol, stigmasta-5,22-dien-3-ol, stigmasterin 9.43 501.1±19.0 °C at 760 mmHg 170 °C Animal study [9], observational human studies [10-12] Cholesterol lowering activity, anti-inflammatory action [13], anti-oxidant action [10]
Beta-Sitosterol [14] C29H50O 414.7

Beta-sitosterol, Cupreol, cinchol, harzol, Rhamnol, Azuprostat, Prostatol, sobatum,

beta-sitosterin

10.482 498 to 501 °C @760 mmHg 143.5 °C Animal studies [15], observational human studies [16-19] Anti-oxidant activity, anti-cancer property [15], mitigation of benign prostatic hyperplasia [18], cholesterol lowering activity [17].
Brassicasterol [20] C28H46O 398.7 Brassicasterin, ergosta-5,22E-dien-3-ol 9.679 488 to 489 °C@760 mmHg 150-151 °C In vitro and in silico studies [21] Anti-infective property, cerebrospinal fluid biomarker for Alzheimer’s disease [22]
Sarsasapogenin [23] C27H44O3 416.6 Parigenin, Sarsagenin, Sarsapogenine 6.210 516.60 °C @760 mmHg 200-201.5 °C In vitro studies [24] Anti-tumor and anti-depressant activity [24], anti-amyloidogenic action in Alzheimer [25]
Cucurbitacin B [26] C32H46O8 558.7 Amarine, Datisca principle B, Cucurbitacin B hydrate 546.74 °C 184-186 °C In vitro and in vivo [27, 28] Anticancer activity [28, 29], inhibits colon cancer [27]
Diosgenin [30] C27H42O3 414.6 Nitogenin, dioscoreasapogenin, 3beta-hydroxy-5-spirostene 6.602 527.11 °C 205.5 °C observational human studies [31-33] Anticancer activity, anti-inflammatory and, anti-infective activity [34], neuroprotective action in Alzheimer’s and Parkinson disease, enhances cognitive function [35]
Ginsenoside Rg1 [36] C42H72O14 801.0 Sanchinoside C1, Panaxoside A, dammarane,-D-glucopyranoside 1.670 902.70 °C @ 760 mmHg 315-318 °C In vitro studies[37] Antidiabetic effect, anti-inflammatory, hepatoprotective and Neuroprotective, Anti-angiopathy[37]
Hecogenin [38] C27H42O4 430.6 Hocogenin 4.22 548.9±50 °C at 760 mmHg 266.5 °C Animal studies [39] Anti-helminthic activity [39], anti-oxidant, anti-inflammatory action and gastro protective action [40]
Jervine [41] C27H39O3 425.6 Iervin, Jerwiny, 11-Ketocyclopamine 3.46 592.5±50 °C at 760 mmHg 243.5-244.5 °C In vitro studies It is teratogen that inhibits smoothened an integral part of hedgehog signalling pathways [42]
Peiminine [43] C27H43NO3 429.6 Verticinone, Fritillarine, Zhebeinone, Raddeanine, Kashmirine, peininine 4.011 567±50 °C at 760 mmHg 212-213 °C In vitro studies [44, 45] Acts as chemo sensitizer for Adriamycin in gastric cancer [44],
Guggulsterone [46, 47] C21H28O2 312.45 Guggulsterones EandZ 3.65 463.3±45 °C at 760 mmHg

E isomer-170-171 °C

Z isomer-188-190 °C

Observational and Animal study [48, 49] Provides cardiovascular protective effect by its hypolipidemic, anti-inflammatory and anti-oxidant action [50], anti-cancer property [49]
Digoxin [51] C41H64O14 780.9 Lanoxin, digacin, davoxin, digosin, cardiogoxin, rougoxin, cordioxil, eudigox, lanacrist, vanoxin 2.37 931.60 °C @760.0 mmHg 230-265 °C Observational studies [52-56] For treatment of atrial fibrillation[54] chronic heart failure [53]
Digitoxin [57] C41H64O13 764.9 Digitoxoside, unidigin, Digitoksin, cardidigin, carditoxin, coramedan, crystodigin, carditalin, digitalin, digitrin 1.85 902± °C at 760 mmHg

493 to 495 °F

256-257 °C (anhydrous)

Observational studies [58-60], In vitro studies and animal study [61] Antiarrhythmic agent [60], treatment for congestive heart failure [58, 59]
Digitonin [62] C56H92O29 1229.3 Digitin 230-240 °C 788.4 °C 446 to 464 °F In vitro studies [63-66]

Biological detergent

[65], induces membrane permeability [63]

Table 2: In silico predicted properties

S. No. Plant steroids Human intestinal absorption CaCo-2 Bioavailability score Ames mutagenesis

Water solubility

Log S

Plasma protein biding (100%) Acute oral toxicity (Kg/mol) Molar refractivity

Lipophilicyity

Log PO/W (WLOGP)

1 Campesterol 0.9930 0.6088 0.585 0.8600

-4.81

Moderately soluble

1.109 3.138 128.42 7.63
2 Ergosterol 0.9950 0.6576 0.542 0.8400 -4.692 moderately soluble 1.018 3.706 127.47 7.33
3 Stigmasterol 0.9914 0.5455 0.5571 0.8300

-4.703

Moderately soluble

1.185 3.285 132.75 7.80
4 Sitosterol 0.9930 0.5385 0.5286 0.8700

-4.703

Poorly soluble

1.124 3.414 133.23 8.02
5 Brassicasterol 0.9914 0.6076 0.5714 0.8700

-4.692

Moderately soluble

1.116 3.195 127.95 7.41
6 Sarsasapogenin 0.9294 0.5138 0.5429 0.8100

-5.182

Moderately soluble

0.941 3.252 122.07 5.79
7 Cucurbitacin B 0.9895 0.7257 0.6571 0.7200 -4.504 Moderately soluble 1.008 5.214 150.94 3.50
8 Diosgenin 0.9482 0.5112 0.6143 0.9100

-6.909

Moderately soluble

1.039 2.97 121.59 5.71
9 Ginsenoside 0.6476 0.8777 0.7571 0.7400 -4.303 0.853 3.993 205.81 1.12
10 Hecogenin 0.9357 0.5126 0.6429 0.7800

-4.59

Moderately soluble

0.774 3.012 122.27 4.97
11 Jervine 0.9789 0.5523 0.5143 0.6700

-3.999

Moderately soluble

0.96 3.327 127.21 3.80
12 Peiminine 0.9638 0.5621 0.5857 0.7000 -2.445 Soluble 1.008 3.332 128.33 3.51
13 Gugglusterone 0.9950 0.8896 0.5714 0.9500 -4.367 0.976 2.922 93.54 4.64
14 Digoxin 0.8851 0.9372 0.7143 0.7400 -4.336 Soluble 0.536 5.056 196.10 2.22
15 Digitoxin 0.8851 0.9257 0.8286 0.7500 -5.066 Soluble 1.055 5.237 194.94 3.25
16 Digitonin -0.7034 0.8742 0.7571 0.7529 -3.414 0.733 5.015 280.34 -6.50


Table 3: Experimental pharmacokinetics

S. No. Phytosteroids AUC Cmax Tmax t1/2 Volume of distribution Clearance Species Route of administration No. of subjects (n) Dose Bioavailability
1 Campesterol [69]

AUC 0-t

120±46.1 ng. h/ml

1250±512 ng/ml 72 (36-72 range) (h) - - - Rats Oral 4 500 mg/kg insadol extract -
2 Ergosterol [70]

AUC (0-36h)

22.29±5.08 (g h ml-1)

22.27±0.19 (g/ml) 8.00±1.18 (h)

5.90±1.41

(h)

- - Rat Oral 6 100 mg/kg -
3 Stigmasterol [69]

AUC (0-∞)

21.8±7.86 ng h/ml

246±38.2 ng/ml

15(9 to72 range)

(h)

- - - Rat Oral 4 500 mg/kg insadol extract -
4 Beta-sitosterol [71]

AUC0-∞

0.19 ng. h/ml

1.8 pg/ml 17.33 (h) 72.4 (h) - - Humans Oral 12 3.8-4.2 µg in 15 ml solution 0.41%
5 Sarsasapogenin [72]

AUC0-48

17405±4398

ng h/ml

2114±362

ng/ml

6 (h) 8.7±2.2 (h) - - Rats Oral 6 25 mg/kg -
6 Cucurbitacin B [73]

AUC0-∞

52.42±29.58

31.24±10.50 µg/l 0.60±0.22 (h) N/A - - Rats Oral 6 4 mg/kg 10.25±5.63 %
7 Diosgenin [74]

AUC0-∞

4.475±0.06 µg h/ml

0.5773±0.012 µg/ml 1 (h) 7.563±0.21 (h) - 2.5143±0.09 ml/h/kg Rats Oral 6 15 mg/kg 9±0.2 %

AUC0-∞

4.957±0.44 µg h/ml

0.6122±0.1 µg/ml - 7.930±0.37 (h) 2.539±0.57 ml/kg 0.222±0.05 ml/h/kg Rats Intravenous single dose 6 1.5 mg/kg -
8 Ginsenoside [75]

AUC0-∞

279.70±81.84 µg/l. h

35.38±10.33 µg/l 4.29±0.76 (h) 3.09±1.64 (h) 848.16±487.89 L/kg 189.36±43.49 L/h/kg Rats Oral 6 50 mg/kg 4.23 %

AUC0-∞

661.31±151.78 µg/l. h

534.97±105.40 µg/l - 6.35±4.04 (h) 69.23±41.13 L/kg

7.88±1.99

L/h/kg

Intravenous 6 5 mg/kg
9 Jervine [76]

AUC0-∞

5547.95±558.34 µg/l. h

233.30±30.37 ng/l 1.20±0.73 (h) 11.09±2.35 (h) 115.24±19.46 L/kg 7.26±0.68 L/h/kg Rats Oral 6 40 mg/kg

AUC0-∞

1289.67±318.46 µg/l. h

138.40±19.31 ng/l 0.13±0.08 (h) 8.35±5.15 (h) 44.15±18.11 L/kg 4.03±0.80 L/h/kg Intravenous 6 5 mg/kg 60.02
10 Peiminine [77]

AUC0-∞

367.2±22.35 ng h/l

68.80±1.63 ng/ml 1.83±0.72 (h) 3.012±0.16 (h) Beagle dog Oral 12 1 g/kg Fritillariaussuriensis powder
11 E-Gugglusterone [78]

AUC0-∞

40.36±6.47 ng. h/ml

0.23±0.01 (h)

41.87±3.95

L/kg

129.06±13.81L/h/kg Rabbit Intravenous 3 10 mg/kg
Z-Guggulsterone

AUC0-∞

20.34±6.2

ng. h/ml

0.35±0.10 (h) 145.09±60.6 L/kg
12 Digoxin [79]

AUC(0-24h)

25.4±6.0 ng. h/ml

AUC(0-∞)

61±13.1 ng. h/ml

3.7±1.7 ng/ml 1.6±1.2 (h) 38±6.1 (h) Human Oral 7 1 mg
13 Digitoxin [80] 138 (h) Human study Oral 6 1 mg 81.5±19.7%
156 (h) 0.47±0.07 L/kg 2.44±0.82 ml/min Intravenous

In silico toxicity prediction holds a great importance in the early drug discovery process as 30-40% of the drug compounds would be rejected at the development stage due to their unidentified toxicities. Ames test is a simple and reliable method to test mutagen city of a compound [35]. It utilises bacterium Salmonella typhimurium strains that carry mutations in genes for histidine synthesis. The lead compound is tested whether it has the ability to cause a reversion to growth on a histidine free medium. The values in the Ames test result have shown their minimal toxicity.

Bai et al., 2018 assessed the in vivo pharmacokinetic (PK) properties of the phytosterol compounds [36, 37]. In this study, male wistar rats of 250-280 gm weight were procured housed in stainless steel cages (3 rats/cage) with room temperature and 12-h light/dark cycle. The rats were fed with standard pellets for a week and were fasted overnight before the commencement of experiments. Pentobarbital (50 mg/kg I. P) to anesthetize the rats, carotid artery cannulated and the blood samples collected in heparin tubes for 0, 15, and 30 min and 1, 2, 4, 6, 8, 10, 12, and 24 h after the oral administration of PSs individually. The blood samples were centrifuged (16,000g, 5 min) and the plasma was separated and stored at 80 °C. The peak plasma concentrations (Cmax) and the time required to reach Cmax (Tmax) of PSs were recorded. The area under the concentration-time curve (AUC) was calculated from the values obtained during 0 to 24 hr using the trapezoidal rule [37]. The half-life (t1/2) was calculated by using the apparent elimination rate constant obtained from the elimination phase gradient. All the data was expressed as mean SD M and the significance was measured by student’s T-test and one-way ANOVA. The significance level was set at p less than 0.05. Using these values, the clearance, volume of distribution and oral bioavailability (BA) were calculated using the non-compartmental analysis (NCA) software PK Solutions version 2.0. All these pharmacokinetic results help the researcher to design the dosing calculations for clinical trials (table 3).

CONCLUSION

In view of the less bioavailability of phytosterols, physical or chemical modification can be applied to increase their bioavailability. Chemical modification has focussed majorly on esterification and physical modification has been attained by microencapsulation method. A physical modification by encapsulation process of phytosterols in nanodelivery system favours dissolution in gastrointestinal tract and enhances the bioavailability. By designing a delivery system, the biologically active substance can be delivered to its specific absorption site. The superiority of this system is that it can decrease the loss of biologically active substances prior to reaching the absorption site to enhance the bioavailability of the compound. The Swiss ADME Web tool used in the study helped in computation of key pharmacokinetic, physicochemical, drug-like and related parameters for one or more molecules. In one attempt, efforts were put in the application to implant free open-access and rapid predictive models exhibiting statistical predictive power, significance, straightforward translation to molecular design and intuitive interpretation,. These methods were adapted from renowned published papers or in-house original methods, especially developed and thoroughly benchmarked. When investigated together, these methods help the researchers to design their studies for improving the bioavailability of phytosterols.

ACKNOWLEDGMENT

The authors are thankful to the Dean, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai and Director, Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh for their support in carrying out this work.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICTS OF INTERESTS

The authors declare no conflict of interest.

REFERENCES

  1. Pendharkar GB, Anjum SD, Patil S. Enhanced biotransformation of phytosterols, a by-product of soybean refineries, to a key intermediate used for synthesis of steroidal drugs. Asian J Pharm Clin Res. 2014;7(5):178-80.

  2. Meng H, Matthan NR, Angellotti E, Pittas AG, Lichtenstein AH. Exploring the effect of vitamin D3 supplementation on surrogate biomarkers of cholesterol absorption and endogenous synthesis in patients with type 2 diabetes-randomized controlled trial. Am J Clin Nutr. 2020;112(3):538-47. doi: 10.1093/ajcn/nqaa149, PMID 32559272.

  3. Sudhop T, Sahin Y, Lindenthal B, Hahn C, Lüers C, Berthold HK. Comparison of the hepatic clearances of campesterol, sitosterol, and cholesterol in healthy subjects suggests that efflux transporters controlling intestinal sterol absorption also regulate biliary secretion. Gut. 2002;51(6):860-3. doi: 10.1136/gut.51.6.860, PMID 12427790.

  4. A Zeez R, S Abaas I, J Kadhim E. Isolation and characterization of β-sitosterol from elaeagnus angustifolia cultivated in Iraq. Asian J Pharm Clin Res 2018;11(11). doi: 10.22159/ajpcr.2018.v11i11.29030.

  5. Le Goff M, Le Ferrec E, Mayer C, Mimouni V, Lagadic Gossmann D, Schoefs B. Microalgal carotenoids and phytosterols regulate biochemical mechanisms involved in human health and disease prevention. Biochimie. 2019;167:106-18. doi: 10.1016/j.biochi.2019.09.012, PMID 31545993.

  6. Lin X, Racette SB, Ma L, Wallendorf M, Spearie CA, Ostlund RE. Plasma biomarker of dietary phytosterol intake. PLOS ONE. 2015;10(2):e0116912. doi: 10.1371/journal.pone.0116912, PMID 25668184.

  7. National Center for Biotechnology Information. PubChem Compound Summary for CID 444679, Ergosterol; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Ergosterol. [Last accessed on 05 Dec 2021]

  8. National Center for Biotechnology Information. PubChem Compound Summary for CID 5280794, Stigmasterol; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Stigmasterol. [Last accessed on 05 Dec 2021]

  9. Elkin RG, Lorenz ES. Feeding laying hens a bioavailable soy sterol mixture fails to enrich their eggs with phytosterols or elicit egg yolk compositional changes. Poult Sci. 2009;88(1):152-8. doi: 10.3382/ps.2008-00271, PMID 19096069.

  10. Hsu CC, Kuo HC, Huang KE. The effects of phytosterols extracted from Diascorea alata on the antioxidant activity, plasma lipids, and hematological profiles in Taiwanese menopausal women. Nutrients. 2017;9(12):1-7. doi: 10.3390/nu9121320, PMID 29206136.

  11. Ho XL, Loke WM. Dietary plant sterols supplementation increases in vivo nitrite and nitrate production in healthy adults: A randomized, controlled study. J Food Sci. 2017;82(7):1750-6. doi: 10.1111/1750-3841.13752, PMID 28708316.

  12. Ras RT, Koppenol WP, Garczarek U, Otten-Hofman A, Fuchs D, Wagner F. Increases in plasma plant sterols stabilize within four weeks of plant sterol intake and are independent of cholesterol metabolism. Nutr Metab Cardiovasc Dis. 2016;26(4):302-9. doi: 10.1016/j.numecd.2015.11.007, PMID 26806045.

  13. Alvarez Sala A, Blanco Morales V, Cilla A, Silvestre RA, Hernandez Alvarez E, Granado Lorencio F. A positive impact on the serum lipid profile and cytokines after the consumption of a plant sterol-enriched beverage with a milk fat globule membrane: a clinical study. Food Funct. 2018;9(10):5209-19. doi: 10.1039/c8fo00353j, PMID 30206618.

  14. National Center for Biotechnology Information. PubChem compound summary for CID 222284, beta-sitosterol; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/beta-sitosterol. [Last accessed on 06 Dec 2021]

  15. Manral C, Roy S, Singh M, Gautam S, Yadav RK, Rawat JK. Effect of β-sitosterol against methyl nitrosourea-induced mammary gland carcinoma in albino rats. BMC Complement Altern Med. 2016;16:260. doi: 10.1186/s12906-016-1243-5, PMID 27473871.

  16. Kasmas SH, Izar MC, França CN, Ramos SC, Moreira FT, Helfenstein T. Differences in synthesis and absorption of cholesterol of two effective lipid-lowering therapies. Braz J Med Biol Res. 2012;45(11):1095-101. doi: 10.1590/s0100-879x2012007500118, PMID 22801416.

  17. Ramprasath VR, Jenkins DJ, Lamarche B, Kendall CW, Faulkner D, Cermakova L. Consumption of a dietary portfolio of cholesterol lowering foods improves blood lipids without affecting concentrations of fat soluble compounds. Nutr J. 2014;13:101. doi: 10.1186/1475-2891-13-101, PMID 25326876.

  18. Sudeep HV, Thomas JV, Shyamprasad K. A double blind, placebo-controlled randomized comparative study on the efficacy of phytosterol-enriched and conventional saw palmetto oil in mitigating benign prostate hyperplasia and androgen deficiency. BMC Urol. 2020;20(1):86. doi: 10.1186/s12894-020-00648-9, PMID 32620155.

  19. Tauriainen MM, Mannisto V, Kaminska D, Vaittinen M, Karja V, Kakela P. Serum, liver and bile sitosterol and sitostanol in obese patients with and without NAFLD. Biosci Rep. 2018;38(2):2-5. doi: 10.1042/BSR20171274, PMID 29540533.

  20. National Center for Biotechnology Information. PubChem compound summary for CID 5281327, Brassicasterol; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/ compound/brassicasterol. [Last accessed on 06 Dec 2021]

  21. Hassan STS. Brassicasterol with dual anti-infective properties against HSV-1 and mycobacterium tuberculosis, and cardiovascular protective effect: nonclinical in vitro and in silico assessments. Biomedicines. 2020;8(5):132. doi: 10.3390/biomedicines8050132, PMID 32456343.

  22. Vanmierlo T, Popp J, Kolsch H, Friedrichs S, Jessen F, Stoffel Wagner B. The plant sterol brassicasterol as additional CSF biomarker in Alzheimer’s disease. Acta psychiatr Scand. 2011;124(3):184-92. doi: 10.1111/j.1600-0447.2011.01713.x, PMID 21585343.

  23. National Center for Biotechnology Information. PubChem compound summary for CID 92095, Sarsasapogenin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/sarsasapogenin. [Last accessed on 06 Dec 2021]

  24. Moon E, Kim AJ, Kim SY. Sarsasapogenin increases melanin synthesis via induction of tyrosinase and microphthalmia-associated transcription factor expression in melan-a cells. Biomol Ther (Seoul). 2012;20(3):340-5. doi: 10.4062/biomolther.2012.20.3.340, PMID 24130933.

  25. Kashyap P, Muthusamy K, Niranjan M, Trikha S, Kumar S. Sarsasapogenin: A steroidal saponin from asparagus racemosus as multi target directed ligand in Alzheimer’s disease. Steroids. 2020;153:108529. doi: 10.1016/j.steroids.2019.108529, PMID 31672628.

  26. National Center for Biotechnology Information. PubChem compound summary for CID 5281316, cucurbitacin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/cucurbitacin-B. [Last accessed on 10 Dec 2021]

  27. Dandawate P, Subramaniam D, Panovich P, Standing D, Krishnamachary B, Kaushik G. Cucurbitacin B and I inhibits colon cancer growth by targeting the Notch signaling pathway. Sci Rep. 2020;10(1):1290. doi: 10.1038/s41598-020-57940-9, PMID 31992775.

  28. Garg S, Kaul SC, Wadhwa R. Cucurbitacin B and cancer intervention: chemistry, biology and mechanisms. Int J Oncol. 2018;52(1):19-37. doi: 10.3892/ijo.2017.4203, PMID 29138804.

  29. Xu N, Zhang BB, Yang MZ, Bai XY, Liang ZQ, Cheng NN. Cucurbitacin B as a Chinese medicine monomer inhibits cell proliferation, invasion, and migration in nasopharyngeal carcinoma. J Nanomater. 2021;2021:1-12. doi: 10.1155/2021/5596780.

  30. National Center for Biotechnology Information. PubChem compound summary for CID 99474, diosgenin 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/diosgenin. [Last accessed on 07 Dec 2021]

  31. Cai T, Cocci A, Cito G, Giammusso B, Zucchi A, Chiancone F. The role of diallyl thiosulfinate associated with nuciferine and diosgenin in the treatment of premature ejaculation: A pilot study. [Archivio Italiano di Urologia, Andrologia: Organo Ufficiale [di] Societa Italiana di Ecografia Urologica e Nefrologica]. 2018;90(1):59-64.

  32. Tohda C, Yang X, Matsui M, Inada Y, Kadomoto E, Nakada S. Diosgenin-rich yam extract enhances cognitive function: A placebo-controlled, randomized, double-blind, crossover study of healthy adults. Nutrients. 2017;9(10):10-6. doi: 10.3390/nu9101160, PMID 29064406.

  33. Yang X, Feng Y, Liu Y, Ye X, Ji X, Sun L. Fuzheng Jiedu Xiaoji formulation inhibits hepatocellular carcinoma progression in patients by targeting the AKT/CyclinD1/p21/p27 pathway. Phytomedicine. 2021;87:153575. doi: 10.1016/j.phymed.2021.153575.

  34. Jesus M, Martins AP, Gallardo E, Silvestre S. Diosgenin: recent highlights on pharmacology and analytical methodology. J Anal Methods Chem. 2016;2016:4156293. doi: 10.1155/2016/4156293, PMID 28116217.

  35. Cai B, Zhang Y, Wang Z, Xu D, Jia Y, Guan Y. Therapeutic potential of diosgenin and its major derivatives against neurological diseases: recent advances. Oxid Med Cell Longev. 2020;2020:3153082. doi: 10.1155/2020/3153082, PMID 32215172.

  36. National Center for Biotechnology Information. PubChem compound summary for CID 3086007, Ginsenosides; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/ compound/ginsenosides. [Last accessed on 07 Dec 2021]

  37. Bai L, Gao J, Wei F, Zhao J, Wang D, Wei J. Therapeutic potential of ginsenosides as an adjuvant treatment for diabetes. Front Pharmacol. 2018;9:423. doi: 10.3389/fphar.2018.00423, PMID 29765322.

  38. National Center for Biotechnology Information. PubChem compound summary for CID 91453, Hecogenin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Hecogenin. [Last accessed on 07 Dec 2021]

  39. Botura MB, Silva GD, Lima HG, Oliveira JV, Souza TS, Santos JD. In vivo anthelmintic activity of an aqueous extract from sisal waste (Agave sisalana Perr.) against gastrointestinal nematodes in goats. Vet Parasitol. 2011;177(1-2):104-10. doi: 10.1016/j.vetpar.2010.11.039, PMID 21156340.

  40. Santos Cerqueira G, Dos Santos, E Silva G, Rios Vasconcelos E, Fragoso de Freitas AP, Arcanjo Moura B, Silveira Macedo D. Effects of hecogenin and its possible mechanism of action on experimental models of gastric ulcer in mice. Eur J Pharmacol. 2012;683(1-3):260-9. doi: 10.1016/j.ejphar.2012.02.043, PMID 22426163.

  41. National Center for Biotechnology Information. PubChem compound summary for CID 10098, Jervine; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Jervine. [Last accessed on 07 Dec 2021]

  42. Chen JK, Taipale J, Cooper MK, Beachy PA. Inhibition of hedgehog signaling by direct binding of cyclopamine to smoothened. Genes Dev. 2002;16(21):2743-8. doi: 10.1101/gad.1025302, PMID 12414725.

  43. National Center for Biotechnology Information. PubChem compound summary for CID 167691, peiminine 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/peiminine. [Last accessed on 07 Dec 2021]

  44. Tang Q, Wang Y, Ma L, Ding M, Li T, Nie Y. Peiminine serves as an adriamycin chemosensitizer in gastric cancer by modulating the EGFR/FAK pathway. Oncol Rep. 2018;39(3):1299-305. doi: 10.3892/or.2018.6184, PMID 29328433.

  45. Zhao B, Shen C, Zheng Z, Wang X, Zhao W, Chen X. Peiminine inhibits glioblastoma in vitro and in vivo through cell cycle arrest and Aautophagic Fflux Bblocking. Cellular Physiology and Biochemistry. 2018;51(4):1566-83. doi: 10.1159/000495646, PMID 30497066.

  46. National Center for Biotechnology Information. PubChem compound summary for CID 6450278, guggulsterone 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/guggulsterone. [Last accessed on 06 Dec 2021]

  47. Sabarathinam S, Rajappan Chandra SK, Thangavel Mahalingam V. CYP3A4 mediated pharmacokinetics drug interaction potential of Maha-Yogaraj Gugglu and E, Z guggulsterone. Scientific Reports. 2021;11(1):715. doi: 10.1038/s41598-020-80595-5, PMID 33436877.

  48. Yamada T, Sugimoto K, Sugimoto K. Guggulsterone and its role in chronic diseases. Advances in Experimental Medicine and Biology. 2016;929:329-61. doi: 10.1007/978-3-319-41342-6_15, PMID 27771932.

  49. Bhat AA, Prabhu KS, Kuttikrishnan S, Krishnankutty R, Babu J, Mohammad RM. Potential therapeutic targets of Guggulsterone in cancer. Nutrition and Metabolism (Lond). 2017;14(1):23. doi: 10.1186/s12986-017-0180-8, PMID 28261317.

  50. Deng R. Therapeutic effects of guggul and its constituent guggulsterone: cardiovascular benefits. Cardiovascular Drug Reviews. 2007;25(4):375-90. doi: 10.1111/j.1527-3466.2007.00023.x, PMID 18078436.

  51. National Center for Biotechnology Information. PubChem compound summary for CID 2724385, Digoxin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/digoxin. [Last accessed on 06 Dec 2021]

  52. Abdul Rahim AH, MacIsaac RL, Jhund PS, Petrie MC, Lees KR, McMurray JJ. Efficacy and safety of digoxin in patients with heart failure and reduced ejection fraction according to diabetes status: an analysis of the Digitalis Investigation Group (DIG) trial. International Journal of Cardiology. 2016;209:310-6. doi: 10.1016/j.ijcard.2016.02.074, PMID 26913372.

  53. Abdul Rahim AH, Shen L, Rush CJ, Jhund PS, Lees KR, McMurray JJV. Effect of digoxin in patients with heart failure and mid-range (borderline) left ventricular ejection fraction. European Journal of Heart Failure. 2018;20(7):1139-45. doi: 10.1002/ejhf.1160, PMID 29493058.

  54. Kotecha D, Bunting KV, Gill SK, Mehta S, Stanbury M, Jones JC. Effect of digoxin vs bisoprolol for heart rate control in atrial fibrillation on patient-reported quality of life: the RATE-AF randomized clinical trial. Jama. 2020;324(24):2497-508. doi: 10.1001/jama.2020.23138, PMID 33351042.

  55. Lam PH, Bhyan P, Arundel C, Dooley DJ, Sheriff HM, Mohammed SF. Digoxin use and lower risk of 30-day all-cause readmission in older patients with heart failure and reduced ejection fraction receiving β-blockers. Clinical Cardiology. 2018;41(3):406-12. doi: 10.1002/clc.22889, PMID 29569405.

  56. Lopes RD, Rordorf R, De Ferrari GM, Leonardi S, Thomas L, Wojdyla DM. Digoxin and mortality in patients with atrial fibrillation. Journal of the American College of Cardiology. 2018;71(10):1063-74. doi: 10.1016/j.jacc.2017.12.060, PMID 29519345.

  57. National Center for Biotechnology Information. PubChem compound summary for CID 441207, Digitoxin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/digitoxin. [Last accessed on 06 Dec 2021].

  58. Bavendiek U, Berliner D, Davila LA, Schwab J, Maier L, Philipp SA. Rationale and design of the DIGIT-HF trial (digitoxin to improve ouTcomes in patients with advanced chronic Heart Failure): a randomized, double-blind, placebo-controlled study. Eur J Heart Fail. 2019;21(5):676-84. doi: 10.1002/ejhf.1452, PMID 30892806.

  59. Holubarsch CJ, Colucci WS, Meinertz T, Gaus W, Tendera M. Survival and prognosis: investigation of crataegus extract WS 1442 in congestive heart failure (SPICE)-rationale, study design and study protocol. European Journal of Heart Failure. 2000;2(4):431-7. doi: 10.1016/s1388-9842(00)00109-4, PMID 11113721.

  60. Ryden L, Hjalmarson AA, Kvasnicka J, Liander B. Haemodynamic effects of the antiarrhythmic quaternary ammonium compound QX-572 in man. British Heart Journal. 1975;37(1):65-73. doi: 10.1136/hrt.37.1.65, PMID 1089426.

  61. Endo H, Yoshida H, Hasegawa M, Ohmi N, Horiuchi N, Hamada Y. Stereoselectivity and species difference in plasma protein binding of KE-298 and its metabolites. Biological and Pharmaceutical Bulletin. 2001;24(7):800-5. doi: 10.1248/bpb.24.800, PMID 11456121.

  62. National Center for Biotechnology Information. PubChem compound summary for CID 6537502, digitonin; 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/ compound/digitonin. [Last accessed on 06 Dec 2021]

  63. Bonventre JV. Calcium in renal cells. Modulation of calcium-dependent activation of phospholipase A2. Environmental Health Perspectives. 1990;84:155-62. doi: 10.1289/ehp.9084155, PMID 2190810.

  64. Lemasters JJ, Holmuhamedov E. Voltage-dependent anion channel (VDAC) as mitochondrial Governator-thinking outside the box. Biochimica et Biophysica Acta. 2006;1762(2):181-90. doi: 10.1016/j.bbadis.2005.10.006, PMID 16307870.

  65. Vonck J, Schafer E. Supramolecular organization of protein complexes in the mitochondrial inner membrane. Biochim Biophys Acta. 2009;1793(1):117-24. doi: 10.1016/j.bbamcr.2008.05.019, PMID 18573282.

  66. Ying M, Chen B, Tian Y, Hou Y, Li Q, Shang X. Nuclear import of human sexual regulator DMRT1 is mediated by importin-beta. Biochimica et Biophysica Acta. 2007;1773(6):804-13. doi: 10.1016/j.bbamcr.2007.03.006, PMID 17459496.

  67. Yang H, Lou C, Sun L, Li J, Cai Y, Wang Z. AdmetSAR 2.0: web-service for prediction and optimization of chemical ADMET properties. Bioinformatics. 2019;35(6):1067-9. doi: 10.1093/bioinformatics/bty707, PMID 30165565.

  68. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports. 2017;7(1):42717. doi: 10.1038/srep42717, PMID 28256516.

  69. Kim D, Park JB, Choi WK, Lee SJ, Lim I, Bae SK. Simultaneous determination of β-sitosterol, campesterol, and stigmasterol in rat plasma by using LC-APCI-MS/MS: application in a pharmacokinetic study of a titrated extract of the unsaponifiable fraction of Zea mays L. Journal of Separation Science. 2016;39(21):4060-70. doi: 10.1002/jssc.201600589, PMID 27591043.

  70. Zhao YY, Cheng XL, Liu R, Ho CC, Wei F, Yan SH. Pharmacokinetics of ergosterol in rats using rapid resolution liquid chromatography-atmospheric pressure chemical ionization multi-stage tandem mass spectrometry and rapid resolution liquid chromatography/tandem mass spectrometry. Journal of Chromatography B, Analytical Technologies in the Biomedical and Life Sciences 2011;879(21):1945-53. doi: 10.1016/j.jchromb.2011.05.025, PMID 21664883.

  71. Duchateau G, Cochrane B, Windebank S, Herudzinska J, Sanghera D, Burian A. Absolute oral bioavailability and metabolic turnover of β-sitosterol in healthy subjects. Drug Metabolism and Disposition: the Biological Fate of Chemical Dispos. 2012;40(10):2026-30. doi: 10.1124/dmd.112.046623, PMID 22826463.

  72. Pei L, Ge S, Ye Y, Jiang Z, Liang X, Zhao W. Development and validation of a UPLC-MS/MS method for determination of sarsasapogenin-AA22 in rat plasma and its application to a pharmacokinetic study. Biomedical Chromatography: BMC. 2018;32(10):e4295. doi: 10.1002/bmc.4295, PMID 29797524.

  73. Hunsakunachai N, Nuengchamnong N, Jiratchariyakul W, Kummalue T, Khemawoot P. Pharmacokinetics of cucurbitacin B from Trichosanthes cucumerina L. in rats. BMC Complementary and Alternative Medicine. 2019;19(1):157-9157. doi: 10.1186/s12906-019-2568-7, PMID 31272429.

  74. Salunkhe R, Gadgoli C, Naik A, Patil N. Pharmacokinetic profile and oral bioavailability of diosgenin, charantin, and hydroxychalcone from a polyherbal formulation. Front Pharmacol. 2021;12:629272-8629272. doi: 10.3389/fphar.2021.629272, PMID 33995027.

  75. Li J, Zhang Y, Fan A, Li G, Liu Q. Pharmacokinetics and bioavailability study of ginsenoside Rk1 in rat by liquid chromatography/electrospray ionization tandem mass spectrometry. Biomedical Chromatography BMC. 2019;33(9):e4580. doi: 10.1002/bmc.4580, PMID 31077415.

  76. Zheng B, Wang C, Song W, Ye X, Xiang Z. Pharmacokinetics and enterohepatic circulation of jervine, an antitumor steroidal alkaloid from Veratrum nigrum in rats. J Pharm Anal. 2019;9(5):367-72. doi: 10.1016/j.jpha.2019.04.004, PMID 31929946.

  77. Wang Z, Cao F, Chen Y, Tang Z, Wang Z. Simultaneous determination and pharmacokinetics of peimine and peiminine in beagle dog plasma by UPLC-MS/MS after the oral administration of ritillariae ussuriensis maxim. and Fritillariae thunbergii Miq. Powder Molecules. 2018;23(7):1573. doi: 10.3390/molecules23071573, PMID 29958456.

  78. Bhatta RS, Kumar D, Chhonker YS, Jain GK. Simultaneous estimation of E- and Z-isomers of guggulsterone in rabbit plasma using liquid chromatography tandem mass spectrometry and its application to pharmacokinetic study. Biomedical Chromatography: BMC. 2011;25(9):1054-60. doi: 10.1002/bmc.1574, PMID 21268049.

  79. Parker RB, Yates CR, Soberman JE, Laizure SC. Effects of grapefruit juice on intestinal P-glycoprotein: evaluation using digoxin in humans. Pharmacotherapy. 2003;23(8):979-87. doi: 10.1592/phco.23.8.979.32881, PMID 12921244.

  80. MacFarland RT, Marcus FI, Fenster PE, Graves PE, Perrier D. Pharmacokinetics and bioavailability of digitoxin by a specific assay. European Journal of Clinical Pharmacology. 1984;27(1):85-9. doi: 10.1007/BF02395212, PMID 6489430.